WO2007028573A1 - Tumor-associated peptides binding to human leukocyte antigen (hla) class i or ii molecules and related anti-cancer vaccine - Google Patents

Tumor-associated peptides binding to human leukocyte antigen (hla) class i or ii molecules and related anti-cancer vaccine Download PDF

Info

Publication number
WO2007028573A1
WO2007028573A1 PCT/EP2006/008641 EP2006008641W WO2007028573A1 WO 2007028573 A1 WO2007028573 A1 WO 2007028573A1 EP 2006008641 W EP2006008641 W EP 2006008641W WO 2007028573 A1 WO2007028573 A1 WO 2007028573A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
peptide
cell
peptides
tumour
Prior art date
Application number
PCT/EP2006/008641
Other languages
English (en)
French (fr)
Other versions
WO2007028573A9 (en
Inventor
Harpreet Singh
Niels Emmerich
Steffen Walter
Toni Weinschenk
Original Assignee
Immatics Biotechnologies Gmbh
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to AU2006289289A priority Critical patent/AU2006289289B2/en
Priority to CA2621414A priority patent/CA2621414C/en
Application filed by Immatics Biotechnologies Gmbh filed Critical Immatics Biotechnologies Gmbh
Priority to DK06777167.5T priority patent/DK1922334T3/da
Priority to CN200680038407.7A priority patent/CN101287755B/zh
Priority to NZ565956A priority patent/NZ565956A/en
Priority to SI200630949T priority patent/SI1922334T1/sl
Priority to KR1020087008126A priority patent/KR101150663B1/ko
Priority to UAA200803781A priority patent/UA97095C2/ru
Priority to EA200800676A priority patent/EA013466B1/ru
Priority to JP2008528437A priority patent/JP5132561B2/ja
Priority to US12/065,725 priority patent/US20090274714A1/en
Priority to BRPI0615466-2A priority patent/BRPI0615466A2/pt
Priority to DE602006019446T priority patent/DE602006019446D1/de
Priority to EP06777167A priority patent/EP1922334B1/en
Priority to PL06777167T priority patent/PL1922334T3/pl
Priority to AT06777167T priority patent/ATE494303T1/de
Publication of WO2007028573A1 publication Critical patent/WO2007028573A1/en
Priority to NO20081690A priority patent/NO20081690L/no
Publication of WO2007028573A9 publication Critical patent/WO2007028573A9/en
Priority to HR20110240T priority patent/HRP20110240T1/xx

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4748Tumour specific antigens; Tumour rejection antigen precursors [TRAP], e.g. MAGE
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators

Definitions

  • HLA human leukocyte antigen
  • the present invention relates to immunotherapeutic methods, and molecules and cells for use in immunotherapeutic methods.
  • the present invention relates to the immunotherapy of cancer, in particular renal cancer.
  • the present invention furthermore relates to tumour-associated T-helper cell peptide epitopes, alone or in combination with other tumour-associated peptides that serve as active pharmaceutical ingredients of vaccine compositions which stimulate anti-tumour immune responses.
  • the present invention relates to two novel peptide sequences derived from HLA class I and II molecules of human tumour cell lines which can be used in vaccine compositions for eliciting anti- tumour immune responses.
  • Stimulation of an immune response is dependent upon the presence of antigens recognised as foreign by the host immune system.
  • tumour associated antigens have now raised the possibility of using a host's immune system to intervene in tumour growth.
  • Various mechanisms of harnessing both the humoral and cellular arms of the immune system are currently being explored for cancer immunotherapy.
  • cytotoxic T-cells CTL
  • CD8-positive T-cells TCD8-positive
  • MHC major histocompatibility complex
  • HLA human leukocyte-antigens
  • MHC-class I- molecules There are two classes of MHC-molecules: MHC-class I- molecules that can be found on most cells having a nucleus that present peptides that result from proteolytic cleavage of endogenous proteins and larger peptides. MHC-class Il-molecules can be found only on professional antigen presenting cells (APC), and present peptides of exogenous proteins that are taken up by APCs during the course of endocytosis, and are subsequently processed.
  • APC professional antigen presenting cells
  • APC professional antigen presenting cells
  • CD8-positive cytotoxic T-lymphocytes complexes of peptide and MHC-II are recognised by CD4-positive -helper- T-cells.
  • CD4-positive helper T-cells play an important role in orchestrating the effector functions of anti-tumour T-cell responses and for this reason the identification of CD4-positive T-cell epitopes derived from tumour associated antigens (TAA) may be of great importance for the development of pharmaceutical products for triggering anti-tumour immune responses (Kobayashi, H., R. Omiya, M. Ruiz, E. Huarte, P. Sarobe, J. J. Lasarte, M. Herraiz, B. Sangro, J. Prieto, F. Borras-Cuesta, and E. Celis. 2002. Identification of an antigenic epitope for helper T lymphocytes from carcinoembryonic antigen. Clin. Cancer Res.
  • CD4 positive T-cells are sufficient for inhibiting manifestation of tumours via inhibition of angiogenesis by secretion of interferon-gamma (IFN ⁇ ) (Qin, Z. and T. Blankenstein. 2000.
  • CD4+ T-cell ⁇ mediated tumour rejection involves inhibition of angiogenesis that is dependent on IFN gamma receptor expression by nonhematopoietic cells. Immunity. 12:677-686).
  • CD4 positive T-cells recognizing peptides from tumour-associated antigens presented by HLA class II molecules can counteract tumour progression via the induction of an Antibody (Ab) responses
  • Ab Antibody
  • a small number of class II ligands of TAA have been described so far (www.cancerimmunity.org, www.syfpeithi.de).
  • HLA class II molecules Since the constitutive expression of HLA class II molecules is usually limited to cells of the immune system (Mach, B., V. Steimle, E. Martinez-Soria, and W. Reith. 1996. Regulation of MHC class II genes: lessons from a disease. Annu. Rev. Immunol. 14:301-331), the possibility of isolating class II peptides directly from primary tumours was not considered possible. Therefore, numerous strategies to target antigens into the class II processing pathway of antigen presenting cells (APCs) have been described, for example the incubation of APCs with the antigen of interest to enable it to be taken up, processed and presented (Chaux, P., V. Vantomme, V. Stroobant, K. Thielemans, J.
  • APCs antigen presenting cells
  • MHC-class-I-binding peptides are usually 8-10 residues in length and contain two conserved residues ("anchors") in their primary amino acid sequence that interact with the corresponding binding groove of the MHC-molecule.
  • MHC class II molecules In the absence of inflammation, expression of MHC class II molecules is mainly restricted to cells of the immune system, especially professional antigen-presenting cells (APC), e.g., monocytes, monocyte-derived cells, macrophages, dendritic cells.
  • APC professional antigen-presenting cells
  • tumour specific T- lymphocytes that is, their epitopes
  • the antigens that are recognised by the tumour specific T- lymphocytes can be molecules derived from all protein classes, such as enzymes, receptors, transcription factors, etc.
  • tumour associated antigens for example, can also be present in tumour cells only, for example as products of mutated genes.
  • tissue-specific structures such as CT ("cancer testis")- antigens that are expressed in different kinds of tumours and in healthy tissue of the testis.
  • CT cancer testis
  • tumour associated antigens have been identified. Further, much research effort is being expended to identify additional tumour associated antigens.
  • Some groups of tumour associated antigens also referred to in the art as tumour specific antigens, are tissue specific.
  • tumour associated antigens identified occur in multiple tumour types, and some, such as oncogenic proteins and/or tumour suppressor genes (tumour suppressor genes are, for example reviewed for renal cancer in Linehan WM, Walther MM, Zbar B. The genetic basis of cancer of the kidney. J Urol. 2003 Dec;170(6 Pt l):2163-72) which actually cause the transformation event, occur in nearly all tumour types.
  • normal cellular proteins that control cell growth and differentiation such as p53 (which is an example for a tumour suppressor gene), ras, c-met, myc, pRB, VHL, and HER-2/neu, can accumulate mutations resulting in upregulation of expression of these gene products thereby making them oncogenic (McCartey et al. Cancer Research 1998 15:58 2601-5; Disis et al. Ciba Found. Symp. 1994 187:198-211).
  • Mucin- 1 is a highly glycosylated type I transmembrane glycoprotein that is abundantly overexpressed on the cell surface of many human adenocarcinomas like breast and ovarian cancers. Aberrant deglycosylation in malignancies is common and unmasks epitopes in tumour cells which might not be presented on normal cells. Moreover, MUCl expression has been demonstrated in multiple myeloma and some B-cell Non-Hodgkin lymphomas (Gendler S, Taylor-Papadimitriou J, Duhig T, Rothbard J, and Burchell J. A highly immunogenic region of a human polymorphic epithelial mucin expressed by carcinomas is made up of tandem repeats. J. Biol.
  • Adipophilin is a marker for specialized differentiated cells containing lipid droplets and for diseases associated with fat-accumulating cells (Heid 1998). Adipophilin occurs in a wide range of cultured cell lines, including fibroblasts and endothelial and epithelial cells. In tissues, however, expression of adipophilin is restricted to certain cell types, such as lactating mammary epithelial cells, adrenal cortex cells, Sertoli and Leydig cells of the male reproductive system, and steatosis or fatty change hepatocytes in alcoholic liver cirrhosis (Heid 1998). Adipophilin has been reported to be overexpressed in colorectal cancer (Saha 2001), hepatocellular carcinoma (Kurokawa 2004), and in renal cell carcinoma (Young 2001).
  • c-Met encodes a heterodimeric transmembranous receptor with tyrosine kinase activity that is composed of an ⁇ -chain that is disulfide-linked to a ⁇ -subunit (Bottaro 1991; Rubin 1993). Both subunits are expressed on the surface, the heavy ⁇ -subunit is responsible for the binding of the ligand, hepatocyte growth factor (HGF), the ⁇ -subunit contains an intracellular domain that mediates the activation of different signal transduction pathways.
  • HGF hepatocyte growth factor
  • c-Met signalling is involved in organ regeneration, as demonstrated for liver and kidney, embryogenesis, haematopoiesis, muscle development, and in the regulation of migration and adhesion of normally activated B-cells and monocytes (Zarnegar 1995; Naldini 1991; Montesano 1998; Schmidt 1995; Uehara 1995; Bladt 1995; Takayama 1996; Mizuno 1993; van, V 1997; Beilmann 2000). Furthermore, numerous studies indicated the involvement of c-Met overexpression in malignant transformation and invasiveness of malignant cells.
  • c-Met mediates the multifunctional and potentially oncogenic activities of the HGF/scatter factor including promotion of cell growth, motility, survival, extracellular matrix dissolution, and angiogenesis (Bottaro 1991; Rubin 1993; Zarnegar 1995). Binding of HGF to the receptor induces autophosphorylation of c-Met and activates downstream signalling events including the ras, phosphatidylinositol 3 '-kinase, phospholipase C ⁇ , and mitogen-activated protein kinase-related pathways (Naldini 1991; Montesano 1998; Furge 2000; Ponzetto 1993; Dong 2001 ; Furge 2001).
  • the c-Met gene is expressed predominantly in epithelial cells and is over- expressed in several malignant tissues and cell lines (Di Renzo 1995; Ferracini 1995; Tuck 1996; Koochekpour 1997; Li 2001; Fischer 1998; Maulik 2002; Qian 2002; Ramirez 2000).
  • An increasing number of reports have shown that nonepithelial cells such as haematopoietic, neural, and skeletal cells respond to HGF and haematological malignancies like multiple myeloma, Hodgkin disease, leukaemia, and lymphoma express the c-Met protein (Gherardi 1991; Teofili 2001; Borset 1999; Jucker 1994; Pons 1998).
  • Regulator of G-Protein Signalling 5 is a negative regulator of heterotrimeric G- protein signalling pathways although its function in vivo remains elusive.
  • RGS proteins comprise a family of molecules with a unifying catalytic function but varying tissue distribution. They stimulate the intrinsic guanosine triphosphatase (GTPase) activity of activated Ga subunits and thereby accelerate G-protein inactivation. Thus, RGS molecules inhibit signalling downstream of G-protein-coupled receptors (De 2000). Recently, it has been shown that Regulator of G-protein signaling-5 induction in pericytes coincides with active vessel remodelling during tumour neovascularization.
  • RGS5 In a mouse model of pancreatic islet cell carcinogenesis, as well as in highly angiogenic astrocytomas, overexpression of RGS 5 has been shown in pericytes during the angiogenic switch accompanying active vessel remodelling. Overexpression was restricted to the tumour vasculature as compared to a normal islet of Langerhans. However, RGS5 is also upregulated during wound healing and ovulation (Berger 2005). Expression of RGS5 is increased in RCC (Rae 2000). In another study, RT-PCR showed strong expression of RGS5 in all RCCs examined, and expression was very weak or undetectable in normal kidneys (6.6:1 by real-time PCR). Tumour endothelial cells were the main location of RGS5 in RCC (Furuya 2004). Furthermore, RGS5 was reported to be a sinusoidal endothelial cell marker in hepatocellular carcinoma (Chen 2004).
  • Apolipoprotein Ll is a secreted high density lipoprotein which binds to apolipoprotein A-I.
  • Apolipoprotein A-I is a relatively abundant plasma protein and is the major apoprotein of HDL. It is involved in the formation of most cholesteryl esters in plasma and also promotes efflux of cholesterol from cells.
  • Apolipoprotein Ll may play a role in lipid exchange and transport throughout the body, as well as in reverse cholesterol transport from peripheral cells to the liver.
  • the plasma protein is a single chain polypeptide with an apparent molecular mass of about 40 kDa (Duchateau 1997; Duchateau 2001).
  • APOLl cDNA was isolated from an activated endothelial cell cDNA library and shown to be upregulated by TNF- ⁇ , which is a potent proinflammatory cytokine. (Monajemi 2002).
  • KIAA0367 was identified in the Kazusa cDNA Project that aims to identify unknown long human transcripts encoding for putative proteins (Ohara 1997). Although the function of the putative 820 amino acid long protein product of KIAA0367 is unknown, it contains a CRAL- TRIO lipid binding domain profile at the C-terminus which binds small hydrophobic molecules and that is present in several nucleotide exchange factors and in the BCL2/adenovirus ElB 19-kDa protein-interacting protein 2 (BNIP-2). BNIP-2 is involved in the control of diverse cellular functions including cell morphology, migration, endocytosis and cell cycle progression (Zhou 2005). KIAA0367 is located on the chromosomal region 9q21. This region is described as a common target of homozygous deletion in many tumours (Gursky 2001; Weber 2001) or loss of heterozygocity (Louhelainen 2000; Tripathi 2003).
  • Soluble guanylate cyclase a heterodimeric protein consisting of an alpha and a beta subunit (1 heme group), catalyzes the conversion of GTP to the second messenger cGMP and functions as the main receptor for nitric oxide and nitrovasodilator drugs (Zabel 1998).
  • GUCYa3 and b3 are overexpressed in human gliomas. Transfection of antisense GUCYl A3 or GUCYl B3 reduced vascularisation and tumour growth in nude mice. This might be due to the fact that VEGF is induced by cGMP (Saino 2004).
  • GUCYl A3 promotes tumour cell migration of a mice mammary tumor cell line (Jadeski 2003).
  • Cyclin Dl belongs to the highly conserved cyclin family, more specific to the cyclin D subfamily (Xiong 1991; Lew 1991). Cyclins function as regulators of CDKs (cyclin- dependent kinases). Different cyclins exhibit distinct expression and degradation patterns which contribute to the temporal coordination of each mitotic event (Deshpande 2005). Cyclin Dl forms a complex with- and functions as a regulatory subunit of CDK4 or CDK6, whose activity is required for cell cycle Gl /S transition. CCNDl forms with CDK4 and CDK6 a serine/threonine kinase holoenzyme complex imparting substrate specificity to the complex (Bates 1994).
  • tumour suppressor protein Rb tumour suppressor protein
  • Rb tumour suppressor protein
  • Mutations, amplification and overexpression of this gene, which alters cell cycle progression, are observed frequently in a variety of tumours and may contribute to tumorigenesis (Hedberg 1999; Vasef 1999; Troussard 2000).
  • MMP7 degrades gelatins, fibronectin and casein (Miyazaki 1990; Quantin 1989) and differs from most MMP family members in that it lacks a conserved C-terminal protein domain (Gaire 1994). MMP7 is often found overexpressed in malignant tissue (Lin 2004; Bramhall 1997; Denys 2004) and it is suggested that it facilitates tumour cell invasion in vivo (Wang 2005).
  • These proteins can be the target of a tumour specific immune response in multiple types of cancer.
  • the Hepatitis B Virus Core Antigen peptide HBV-001 is not derived from an endogenous human tumour-associated antigen, but is derived from the Hepatitis B virus core antigen. Firstly, it allows to quantitavely compare the magnitude of T-cell responses induced by TUMAPs and hence allows important conclusions on the capacity to elicit anti-tumour responses. Secondly, it functions as an important positive control in the case of lack of any T- cell responses in the patient. And thirdly, it also allows to conclude on the status of immunocompetence of the patient.
  • HBV Hepatitiv B virus
  • the HBV genome (Previsani 2002) is comprised of partially double-stranded circular DNA. In HBV virions, it is packed together with the core protein HBc and other proteins to form the nucleocapsid, which is surrounded by an outer envelope containing lipids and the surface protein family HBs (also called envelope protein).
  • HBcAg and HBsAg The antigenic determinants which are associated with HBc and HBs are noted as HBcAg and HBsAg, respectively. These antigens are associated with serological, i.e. antibody responses found in the patient blood and are among the clinically most useful antigen-antibody systems for the diagnosis of HBV infection.
  • HBc will represent a novel foreign antigen for all individuals without prior history of HBV infection.
  • immunogenic peptides are well known for this antigen (Bertoletti 1993; Livingston 1997), one ten-amino acid peptide from HBcAg was selected as a positive control antigen within IMA. The induction of HBc peptide-specific CTLs will then be used as a marker for patient immunocompetence and successful vaccination.
  • Immunotherapy in cancer patients aims at activating cells of the immune system specifically, especially the so-called cytotoxic T-cells (CTL, also known as “killer cells”, also known as CD8-positive T-cells), against tumour cells but not against healthy tissue.
  • CTL cytotoxic T-cells
  • Tumour cells differ from healthy cells by the expression of tumour-associated proteins.
  • HLA molecules on the cell surface present the cellular content to the outside, thus enabling a cytotoxic T-cell to differentiate between a healthy and a tumour cell. This is realized by breaking down all proteins inside the cell into short peptides, which are then attached to HLA molecules and presented on the cell surface (Rammensee 1993).
  • Peptides that are presented on tumour cells, but not or to a far lesser extent on healthy cells of the body are called tumour-associated peptides (TUMAPs).
  • tumour-specific antigen In order for the proteins to be recognised by the cytotoxic T-lymphocytes as tumour-specific antigen, and in order to be used in a therapy, particular prerequisites must be fulfilled.
  • the antigen should be expressed mainly by tumour cells and not by normal healthy tissues or in rather small amounts. It is furthermore desirable, that the respective antigen is not only present in one type of tumour, but also in high concentrations (e.g. copy numbers per cell).
  • Essential is the presence of epitopes in the amino acid sequence of the antigen, since such peptide ("immunogenic peptide") that is derived from a tumour associated antigen should lead to an in vitro or in vivo T-cell-response.
  • Tl ⁇ 7.0 cm, limited to the kidney Nl single regional lymph node T2 > 7.0 cm, limited to the kidney N2 more than one regional lymph node
  • the standard treatment for RCC is radical nephrectomy (for all stages). Radiation therapy may be used to reduce the cancer's spread, but renal cell carcinomas are often resistant to radiation. Hormonal therapy may reduce the growth of the tumour in some cases (less than 10%). To date chemotherapy has not demonstrated any significant activity in this disease. Vinblastine, 5-FU (5-fluorouracil) and floxuridane (FUDR) are the chemotherapy drugs that have been studied most, but only 5-FU and its metabolite FUDR have demonstrated a 10-12% activity rate (Vokes EE, and Golomb HM. Oncologic Therapies, 2nd edition. Springer- Verlag, Berlin/Heidelberg (2003)). The combination of gemcitabine and 5-FU resulted in a 17% response rate.
  • 5-FU 5-fluorouracil
  • FUDR floxuridane
  • Immunological treatments such as Interferon alpha (IFN ⁇ ) or Interleukin-2 (IL-2) have been evaluated in recent years by regulatory authorities in the USA and Europe for the treatment of advanced RCC.
  • High dose IL-2 treatment is up to date still the only immunological regimen being approved by the FDA.
  • IFN ⁇ monotherapy was initially reported to have a 25-30% response rate but many additional trials have suggested a true response rate of only about 10% (Vokes EE, and Golomb HM. Oncologic Therapies, 2nd edition. Springer- Verlag, Berlin/Heidelberg (2003)).
  • IL-2 appears to have a similar overall response rate compared to IFN ⁇ with approximately 5% of the patients achieving durable complete remissions (Rosenberg 1987).
  • T-helper cells play an important role in orchestrating the effector function of CTLs in anti- tumour immunity.
  • T-helper cell epitopes that trigger a T-helper cell response of the THl type support effector functions of CD8-positive Killer T-cells, which include cytotoxic functions directed against tumour cells displaying tumour-associated peptide/MHC complexes on their cell surfaces.
  • tumour-associated T-helper cell peptide epitopes alone or in combination with other tumour-associated peptides, can serve as active pharmaceutical ingredients of vaccine compositions which stimulate anti -tumour immune responses.
  • tumour vaccine The major task in the development of a tumour vaccine is therefore the identification and characterisation of novel tumour associated antigens and immunogenic T-helper epitopes derived thereof, that can be recognised by CD8-positive T-cells, or CD4-positive T-cells, in particular CD4-positive T-cells of the Tm type. It is therefore an object of the present invention to provide novel amino acid sequences for such peptides that have the ability to bind to a molecule of the human major histocompatibility complex (MHC) class-I (HLA class I) or II (HLA class II). It is a further object of the present invention, to provide an effective anti-cancer vaccine that is, at least in part, based on said novel peptides.
  • MHC human major histocompatibility complex
  • the first object is solved by providing a tumour associated peptide that is selected from the group of peptides comprising at least one sequence according to any of SEQ ID No. 1 or SEQ ID No. 2 of the attached sequence listing, wherein one peptide has the ability to bind to a molecule of the human major histocompatibility complex (MHC) class-II (HLA class II), and the other has the ability to bind to a molecule of the human major histocompatibility complex (MHC) class-I (HLA class I), provided that the peptide is not the intact human tumour associated polypeptide.
  • MHC human major histocompatibility complex
  • HLA class II human major histocompatibility complex
  • HLA class I human major histocompatibility complex
  • the inventors demonstrate that it is possible to isolate and characterize peptides binding to HLA class I or II molecules directly from mammalian tumours, preferentially human tumours, preferably renal cell carcinomas.
  • the present invention provides peptides that stem from antigens associated with tumourigenesis, and have the ability to bind sufficiently to HLA class II molecules for triggering an immune response of human leukocytes, especially lymphocytes, especially T lymphocytes, especially CD4-positive T lymphocytes, especially CD4-positive T lymphocytes mediating Tm-type immune responses.
  • the present invention also provides peptides that stem from antigens associated with tumourigenesis, and have the ability to bind sufficiently to HLA class I molecules for triggering an immune response of human leukocytes, especially lymphocytes, especially T lymphocytes, especially CD8-positive cytotoxic T-lymphocytes.
  • tumour-associated antigens especially tumour-associated antigens with functions in, e.g., proteolysis, angiogenesis, cell growth, cell cycle regulation, cell division, regulation of transcription, regulation of translation, tissue invasion, including, e.g., tumour-associated peptides from matrix-metalloproteinase 7 (MMP7; SEQ ID No. 1) and Apolipoprotein Ll (APOLl; SEQ ID No. 4).
  • MMP7 matrix-metalloproteinase 7
  • Apolipoprotein Ll Apolipoprotein Ll
  • tumour-associated peptides sufficiently binding promiscuously to HLA-class II molecules, especially those HLA class II alleles genetically encoded by HLA DR loci of the human genome, are able to elicit immune responses mediated by human CD4-positive T-cells.
  • CD4-positive T-cells were isolated from human peripheral blood, demonstrating that the claimed peptides are suitable for triggering T-cell responses of the human immune system against selected peptides of the tumour cell peptidome. As exemplified below with a peptide from MMP7 (SEQ ID No. 1), this promiscuously HLA-DR-binding, tumour-associated peptide was found to be recognized by CD4-positive T-cells.
  • tumour-associated peptides sufficiently binding to HLA-class I molecules are able to elicit immune responses mediated by human CD8-positive cytotoxic T- lymphocytes, also demonstrating that the claimed peptides are suitable for triggering responses of the human immune system against selected peptides of the tumour cell peptidome.
  • peptides can be synthesized chemically and can be used as active pharmaceutical ingredients of pharmaceutical preparations, the peptides provided by the present invention can be used for immunotherapy, preferentially cancer immunotherapy.
  • the second object of the present invention is solved by providing a pharmaceutical preparation, preferably in the form of a vaccine, that is effective against cancer cells, in particular cells of solid tumours, comprising an effective amount of a peptide according to the invention, or comprising a nucleic acid encoding such a peptide.
  • a pharmaceutical preparation preferably in the form of a vaccine, that is effective against cancer cells, in particular cells of solid tumours, comprising an effective amount of a peptide according to the invention, or comprising a nucleic acid encoding such a peptide.
  • the vaccine can furthermore contain additional peptides and/or excipients to be more effective, as will be further explained below.
  • the peptide or peptide-encoding nucleic acid can also constitute a tumour or cancer vaccine. It may be administered directly into the patient, into the affected organ or systemically, or applied ex vivo to cells derived from the patient or a human cell line which are subsequently administered to the patient, or used in vitro to select a subpopulation from immune cells derived from the patient, which are then re-administered to the patient.
  • a peptide comprising an amino acid sequence according to SEQ ID No. 1 (SQDDIKGIQKL YGKRS) or SEQ ID No. 2 (VMAGDIYSV) or a variant thereof, provided that the peptide is not the intact human polypeptide from which the amino acid sequence is derived (i.e. one of the full-length sequences as listed in the locus link IDs (Accession numbers, see the attached Table 1, below).
  • peptides that form the basis of the present invention have all been identified as being presented by MHC class I or II bearing cells (RCC).
  • RRC MHC class I or II bearing cells
  • these particular peptides as well as other peptides containing the sequence (i.e. derived peptides) all elicit a specific T-cell response, although the extent to which such response will be induced might vary from individual peptide to peptide. Differences, for example, could be caused due to mutations in said peptides (see below).
  • the person of skill in the present art is well aware of methods that can be applied in order to determine the extent to which a response is induced by an individual peptide, in particular with reference to the examples herein and the respective literature.
  • a peptide according to the present invention consists essentially of an amino acid sequence according to SEQ ID No. 1 or SEQ ID No. 2 or a variant thereof.
  • Consisting essentially of shall mean that a peptide according to the present invention, in addition to the sequence according to any of SEQ ID No. 1 to SEQ ID No. 11 or a variant thereof, contains additional N- and/or C-terminally located stretches of amino acids that are not necessarily forming part of the peptide that functions as core sequence of the peptide comprising the binding motif and as an immunogenic T-helper epitope.
  • the peptide of the present invention comprises the 80 N-terminal amino acids of the HLA-DR antigen-associated invariant chain (p33, in the following "Ii") as derived from the NCBI, GenBank Accession-number X00497 (Strubin, M., Mach, B. and Long, E.O.
  • the complete sequence of the mRNA for the HLA-DR-associated invariant chain reveals a polypeptide with an unusual transmembrane polarity EMBO J. 3 (4), 869-872 (1984)).
  • a “variant" of the given amino acid sequence we mean that the side chains of, for example, one or two of the amino acid residues are altered (for example by replacing them with the side chain of another naturally occurring amino acid residue or some other side chain) such that the peptide is still able to bind to an HLA molecule in substantially the same way as a peptide consisting of the given amino acid sequence.
  • a peptide may be modified so that it at least maintains, if not improves, the ability to interact with and bind to a suitable MHC molecule, such as HLA-DRBl in the case of HLA class II molecules, or HLA- A2 in the case of class I molecules, and so that it at least maintains, if not improves, either the ability to generate activated CTL which can recognise and kill cells which aberrantly express a polypeptide which contains an amino acid sequence as defined in the aspects of the invention, or the ability to stimulate helper T-cells which can provide help to CD8 positive T-cells or directly attack target cells by secreting cytokines.
  • a suitable MHC molecule such as HLA-DRBl in the case of HLA class II molecules, or HLA- A2 in the case of class I molecules
  • HLA-DR binding peptides are typically anchor residues forming a core sequence fitting to the binding motif of the HLA binding groove. Modifications of these and other residues involved in binding HLA-DR may enhance binding without altering CTL recognition. Those amino acid residues that are not essential to interact with the T-cell receptor can be modified by replacement with another amino acid whose incorporation does not substantially affect T-cell reactivity and does not eliminate binding to the relevant MHC.
  • the peptide of the invention may be any peptide (by which term we include oligopeptide or polypeptide) which includes the amino acid sequences or a portion or variant thereof as given.
  • a preferred peptide of the present invention exhibits an overall length of between 9 and 100, preferably between 9 and 30, and most preferred between 9 and 16 amino acids.
  • peptides can be used either directly in order to load MHC class II molecules or the sequence can be cloned into the vectors according to the description herein below. As these peptides form the final product of the processing of larger peptides within the cell, longer peptides can be used as well.
  • the peptides of the invention may be of any size, but typically they may be less than 100,000 Da in molecular weight, preferably less than 50,000 Da, more preferably less than 10,000 Da and typically about 5,000 Da. In terms of the number of amino acid residues, the peptides of the invention may have fewer than 1000 residues, preferably fewer than 500 residues, more preferably fewer than 100 residues.
  • the peptides of the invention may be used to trigger an MHC class I specific response, as the peptides can exhibit simultaneous core- or partial sequences of HLA class I-molecules.
  • a preferred MHC class I specific peptide of the present invention exhibits an overall length of between 9 and 16, preferably between 9 and 12 amino acids. It shall be understood that those peptides might be used (for example in a vaccine) as longer peptides, similar to MHC class II peptides.
  • the peptides of the invention are particularly useful in immunotherapeutic methods for enabling T-cells to recognize cells which aberrantly express polypeptides that form the basis for the present peptides of the invention. Since these specific peptides consisting of the given amino acid sequences bind to HLA class I or HLA class II molecules it is preferred that the peptides of the invention are ones which bind HLA class I or HLA class II molecules and when so bound the HLA-peptide complex is present on the surface of a suitable antigen- presenting cell, is capable of eliciting the stimulation of T-cells which recognise cells which aberrantly express a polypeptide comprising the given amino acid sequence.
  • a peptide which is greater than around 12 amino acid residues is used directly to bind to a MHC molecule, it is preferred that the residues that flank the core HLA binding region are ones that do not substantially affect the ability of the peptide to bind specifically to the binding groove of the MHC molecule or to present the peptide to the T-cells.
  • the residues that flank the core HLA binding region are ones that do not substantially affect the ability of the peptide to bind specifically to the binding groove of the MHC molecule or to present the peptide to the T-cells.
  • larger peptides may be used, especially when encoded by a polynucleotide, since these larger peptides may be fragmented by suitable antigen-presenting cells.
  • Examples for peptides of MHC ligands, motifs, variants, as well as certain examples for N- and/or C-terminal extensions can be, for example, derived from the database SYFPEITHI (Rammensee H, Bachmann J, Emmerich NP, Bachor OA, Stevanovic S. SYFPEITHI: database for MHC ligands and peptide motifs. Immunogenetics.1999 Nov; 50(3-4):213-9. Review.) at http://syfpeithi.bmi-heidelberg.com/ and the references as cited therein.
  • certain peptides for HLA-DR in the database are K H K V YAC EVTHQG LSS derived from Ig kappa chain 188-203 (Kovats et al. Eur J Immunol.1997 Apr;27(4):1014-21); KVQ WKVDNALOS GNS derived from Ig kappa chain 145- 159 (Kovats et al. Eur J Immunol.1997 Apr;27(4): 1014-21), LPRLI AFTSEHSHF derived from GAD65270-283 (Endl et al.
  • peptides can also be derived from mutated sequences of antigens, such as in the case of ATGFKQSSKA LQRPVAS derived from bcr-abl 210 kD fusion protein (ten Bosch et al. Blood. 1996 Nov l;88(9):3522-7), G Y K V L V L N P S V A A T derived from HCV-I NS3 28-41 Diepolder et al. J Virol.
  • peptide the inventors include not only molecules in which amino acid residues are joined by peptide (-CO-NH-) linkages but also molecules in which the peptide bond is reversed.
  • retro-inverso peptidomimetics may be made using methods known in the art, for example such as those described in Meziere et al (1997) J. Immunol. 159,3230-3237, incorporated herein by reference. This approach involves making pseudopeptides containing changes involving the backbone, and not the orientation of side chains. Meziere et al (1997) show that, at least for MHC class II and T helper cell responses, these pseudopeptides are useful.
  • Retro-inverse peptides which contain NH-CO bonds instead of CO-NH peptide bonds, are much more resistant to proteolysis.
  • the peptide of the invention is one which, if expressed in an antigen presenting cell, may be processed so that a fragment is produced which is able to bind to an appropriate MHC molecule and may be presented by a suitable cell and elicit a suitable T-cell response. It will be appreciated that a fragment produced from the peptide may also be a peptide of the invention.
  • the peptide of the invention contains a portion which includes the given amino acid sequence or a portion or variant thereof and a further portion which confers some desirable property.
  • the further portion may include a further T-cell epitope (whether or not derived from the same polypeptide as the first T-cell epitope-containing portion) or it may include a carrier protein or peptide.
  • the peptide of the invention is a truncated human protein or a fusion protein of a protein fragment and another polypeptide portion provided that the human portion includes one or more inventive amino acid sequences.
  • the peptides of the invention include the amino acid sequences of the invention and at least one further T-cell epitope wherein the further T-cell epitope is able to facilitate the production of a T-cell response directed at the type of tumour that aberrantly expresses a tumour-associated antigen.
  • the peptides of the invention include so-called "beads on a string" polypeptides which can also be used as vaccines.
  • Such peptides can be spaced apart by chemical linkers, which might contain amino acids (such as G-stretches), but which can - additionally or alternatively - comprise chemical linking groups (i.e. not having a function except for providing a particular spacing).
  • the polypeptide is over-expressed compared to normal levels of expression or that the gene is silent in the tissue from which the tumour is derived but in the tumour it is expressed.
  • over-expressed we mean that the polypeptide is present at a level at least 1.2 x that present in normal tissue; preferably at least 2 x and more preferably at least 5 x or 10 x the level present in normal tissue.
  • Peptides (at least those containing peptide linkages between amino acid residues) may be synthesised by the Fmoc-polyamide mode of solid-phase peptide synthesis as disclosed by Lu et al (1981) J. Org. Chem. 46, 3433-3436, and references therein.
  • Temporary N-amino group protection is afforded by the 9-fluorenylmethyloxycarbonyl (Fmoc) group. Repetitive cleavage of this highly base-labile protecting group is achieved by using 20 % piperidine in N, N-dimethylformamide.
  • Side-chain functionalities may be protected as their butyl ethers (in the case of serine threonine and tyrosine), butyl esters (in the case of glutamic acid and aspartic acid), butyloxycarbonyl derivative (in the case of lysine and histidine), trityl derivative (in the case of cysteine) and 4-rnethoxy-2,3,6-trimethylbenzenesulphonyl derivative (in the case of arginine).
  • glutamine or asparagine are C-terminal residues, use is made of the 4,4'-dimethoxybenzhydryl group for protection of the side chain amido functionalities.
  • the solid-phase support is based on a polydimethyl-acrylamide polymer constituted from the three monomers dimethylacrylamide (backbone-monomer), bisacryloylethylene diamine (cross linker) and acryloylsarcosine methyl ester (functionalising agent).
  • the peptide-to-resin cleavable linked agent used is the acid-labile 4-hydroxymethyl- phenoxyacetic acid derivative. All amino acid derivatives are added as their preformed symmetrical anhydride derivatives with the exception of asparagine and glutamine, which are added using a reversed N, N-dicyclohexyl-carbodiimide/lhydroxybenzotriazole mediated coupling procedure.
  • Trifluoroacetic acid is removed by evaporation in vacuo, with subsequent trituration with diethyl ether affording the crude peptide.
  • Any scavengers present are removed by a simple extraction procedure which on lyophilisation of the aqueous phase affords the crude peptide free of scavengers.
  • Reagents for peptide synthesis are generally available from Calbiochem- Novabiochem (UK) Ltd, Nottingham NG7 2QJ, UK.
  • Purification may be effected by any one, or a combination of, techniques such as size exclusion chromatography, ion-exchange chromatography and (usually) reverse-phase high performance liquid chromatography.
  • Analysis of peptides may be carried out using thin layer chromatography, reverse-phase high performance liquid chromatography, amino-acid analysis after acid hydrolysis and by fast atom bombardment (FAB) mass spectrometric analysis, as well as MALDI and ESI-Q-TOF mass spectrometric analysis.
  • FAB fast atom bombardment
  • a further aspect of the invention provides a nucleic acid (e.g. polynucleotide) encoding a peptide of the invention.
  • the nucleic acid according to the present invention may be DNA, cDNA, PNA, CNA, RNA or combinations thereof and it may or may not contain introns as long as it codes for the peptide.
  • a still further aspect of the invention provides an expression vector capable of expressing a polypeptide according to the invention.
  • a variety of methods have been developed to operably link polynucleotides, especially DNA, to vectors for example via complementary cohesive termini. For instance, complementary homopolymer tracts can be added to the DNA segment to be inserted to the vector DNA. The vector and DNA segment are then joined by hydrogen bonding between the complementary homopolymeric tails to form recombinant DNA molecules.
  • Synthetic linkers containing one or more restriction sites provide an alternative method of joining the DNA segment to vectors.
  • the DNA segment generated by endonuclease restriction digestion as described earlier, is treated with bacteriophage T4 DNA polymerase or E. coli DNA polymerase I, enzymes that remove protruding, 3 '-single-stranded termini with their 3'-5'-exonucleolytic activities, and fill in recessed 3 '-ends with their polymerising activities.
  • the combination of these activities therefore generates blunt-ended DNA segments.
  • the blunt-ended segments are then incubated with a large molar excess of linker molecules in the presence of an enzyme that is able to catalyse the ligation of blunt-ended DNA molecules, such as bacteriophage T4 DNA ligase.
  • an enzyme that is able to catalyse the ligation of blunt-ended DNA molecules, such as bacteriophage T4 DNA ligase.
  • the products of the reaction are DNA segments carrying polymeric linker sequences at their ends.
  • These DNA segments are then cleaved with the appropriate restriction enzyme and ligated to an expression vector that has been cleaved with an enzyme that produces termini compatible with those of the DNA segment.
  • Synthetic linkers containing a variety of restriction endonuclease sites are commercially available from a number of sources including International Biotechnologies Inc, New Haven, CN, USA.
  • a desirable way to modify the DNA encoding the polypeptide of the invention is to use the polymerase chain reaction as disclosed by Saiki et al (1988) Science 239,487-491.
  • This method may be used for introducing the DNA into a suitable vector, for example by engineering in suitable restriction sites, or it may be used to modify the DNA in other useful ways as is known in the art.
  • the DNA to be enzymatically amplified is flanked by two specific primers which themselves become incorporated into the amplified DNA.
  • the said specific primers may contain restriction endonuclease recognition sites which can be used for cloning into expression vectors using methods known in the art.
  • the DNA (or in the case of retroviral vectors, RNA) is then expressed in a suitable host to produce a polypeptide comprising the compound of the invention.
  • the DNA encoding the polypeptide constituting the compound of the invention may be used in accordance with known techniques, appropriately modified in view of the teachings contained herein, to construct an expression vector, which is then used to transform an appropriate host cell for the expression and production of the polypeptide of the invention.
  • Such techniques include those disclosed in US Patent Nos.
  • DNA (or in the case of retroviral vectors, RNA) encoding the polypeptide constituting the compound of the invention may be joined to a wide variety of other DNA sequences for introduction into an appropriate host.
  • the companion DNA will depend upon the nature of the host, the manner of the introduction of the DNA into the host, and whether episomal maintenance or integration is desired.
  • the DNA is inserted into an expression vector, such as a plasmid, in proper orientation and correct reading frame for expression.
  • an expression vector such as a plasmid
  • the DNA may be linked to the appropriate transcriptional and translational regulatory control nucleotide sequences recognised by the desired host, although such controls are generally available in the expression vector.
  • the vector is then introduced into the host through standard techniques. Generally, not all of the hosts will be transformed by the vector. Therefore, it will be necessary to select for transformed host cells.
  • One selection technique involves incorporating into the expression vector a DNA sequence, with any necessary control elements, that codes for a selectable trait in the transformed cell, such as antibiotic resistance.
  • the gene for such selectable trait can be on another vector, which is used to co- transform the desired host cell.
  • Host cells that have been transformed by the recombinant DNA of the invention are then cultured for a sufficient time and under appropriate conditions known to those skilled in the art in view of the teachings disclosed herein to permit the expression of the polypeptide, which can then be recovered.
  • Many expression systems are known, including bacteria (for example E. coli and Bacillus subtilis), yeasts (for example Saccharomyces cerevisiae), filamentous fungi (for example Aspergillus), plant cells, animal cells and insect cells.
  • the system can be RCC or Awells cells.
  • a promoter is an expression control element formed by a DNA sequence that permits binding of RNA polymerase and transcription to occur.
  • Promoter sequences compatible with exemplary bacterial hosts are typically provided in plasmid vectors containing convenient restriction sites for insertion of a DNA segment of the present invention.
  • Typical prokaryotic vector plasmids are pUC18, pUC19, pBR322 and pBR329 available from Biorad Laboratories, (Richmond, CA, USA) and pTrc99A and pKK223-3 available from Pharmacia, Piscataway, NJ, USA.
  • a typical mammalian cell vector plasmid is pSVL available from Pharmacia, Piscataway, NJ, USA. This vector uses the SV40 late promoter to drive expression of cloned genes, the highest level of expression being found in T antigen-producing cells, such as COS-I cells.
  • An example of an inducible mammalian expression vector is pMSG, also available from Pharmacia. This vector uses the glucocorticoid-inducible promoter of the mouse mammary tumour virus long terminal repeat to drive expression of the cloned gene.
  • Useful yeast plasmid vectors are pRS403-406 and pRS413-416 and are generally available from Stratagene Cloning Systems, La Jolla, CA 92037, USA.
  • Plasmids pRS403, pRS404, pRS405 and pRS406 are Yeast Integrating plasmids (Yips) and incorporate the yeast selectable markers HIS3, TRPl, LEU2 and URA3.
  • Plasmids pRS413-416 are Yeast Centromere plasmids (Ycps). Other vectors and expression systems are well known in the art for use with a variety of host cells.
  • the present invention also relates to a host cell transformed with a polynucleotide vector construct of the present invention.
  • the host cell can be either prokaryotic or eukaryotic.
  • Bacterial cells may be preferred prokaryotic host cells in some circumstances and typically are a strain of E. coli such as, for example, the E. coli strains DH5 available from Bethesda Research Laboratories Inc., Bethesda, MD, USA, and RRl available from the American Type Culture Collection (ATCC) of Rockville, MD, USA (No ATCC 31343).
  • ATCC American Type Culture Collection
  • Preferred eukaryotic host cells include yeast, insect and mammalian cells, preferably vertebrate cells such as those from a mouse, rat, monkey or human fibroblastic and kidney cell lines.
  • Yeast host cells include YPH499, YPH500 and YPH501 which are generally available from Stratagene Cloning Systems, La Jolla, CA 92037, USA.
  • Preferred mammalian host cells include Chinese hamster ovary (CHO) cells available from the ATCC as CCL61, NIH Swiss mouse embryo cells NIH/3T3 available from the ATCC as CRL 1658, monkey kidney-derived COS-I cells available from the ATCC as CRL 1650 and 293 cells which are human embryonic kidney cells.
  • Preferred insect cells are Sf9 cells which can be transfected with baculovirus expression vectors.
  • Transformation of appropriate cell hosts with a DNA construct of the present invention is accomplished by well known methods that typically depend on the type of vector used.
  • transformation of prokaryotic host cells see, for example, Cohen et al (1972) Proc. Natl. Acad. Sci. USA 69,2110 and Sambrook et al (1989) Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY. Transformation of yeast cells is described in Sherman et al (1986) Methods In Yeast Genetics, A Laboratory Manual, Cold Spring Harbor, NY. The method of Beggs (1978) Nature 275,104-109 is also useful.
  • reagents useful in transfecting such cells for example calcium phosphate and DEAE-dextran or liposome formulations, are available from Stratagene Cloning Systems, or Life Technologies Inc., Gaithersburg, MD 20877, USA. Electroporation is also useful for transforming and/or transfecting cells and is well known in the art for transforming yeast cell, bacterial cells, insect cells and vertebrate cells.
  • Successfully transformed cells i.e. cells that contain a DNA construct of the present invention
  • cells resulting from the introduction of an expression construct of the present invention can be grown to produce the polypeptide of the invention.
  • Cells can be harvested and lysed and their DNA content examined for the presence of the DNA using a method such as that described by Southern (1975) J. MoI. Biol. 98,503 or Berent et al (1985) Biotech. 3,208.
  • the presence of the protein in the supernatant can be detected using antibodies as described below.
  • the present invention also contemplates a culture of those cells, preferably a monoclonal (clonally homogeneous) culture, or a culture derived from a monoclonal culture, in a nutrient medium.
  • host cells of the invention are useful in the preparation of the peptides of the invention, for example bacterial, yeast and insect cells.
  • other host cells may be useful in certain therapeutic methods.
  • antigen-presenting cells such as dendritic cells, may usefully be used to express the peptides of the invention such that they may be loaded into appropriate MHC molecules.
  • Preferred host cells are recombinant RCC or Awells cells.
  • Preferred is a method of producing a tumour associated peptide according to the present invention, the method comprising culturing the host cell according to the present invention, and isolating the peptide from the host cell or its culture medium, according to standard methods.
  • a further aspect of the invention provides a method of producing a peptide for oral, rectal, nasal or lingual uptake, intravenous (i.v.) injection, sub-cutaneous (s.c.) injection, intradermal (i.d.) injection, intraperitoneal (i.p.) injection, intramuscular (i.m.) injection.
  • intravenous i.v.
  • sub-cutaneous s.c.
  • intradermal i.d.
  • intraperitoneal injection i.p.
  • intramuscular injection i.m.
  • Preferred ways of peptide injection are s.c, i.d., i.p., i.m., and i.v.
  • Preferred ways of DNA injection are i.d., i.m., s.c, i.p. and i.v.
  • Doses of between 0.1 and 500 mg of peptide or DNA may be given, as is also outlined below.
  • a further aspect of the invention relates to the use of a tumour associated peptide according to the invention, a nucleic acid according to the invention or an expression vector according to the invention in medicine.
  • the object of the present invention in a further aspect thereof, is solved by a pharmaceutical composition that contains at least one tumour associated peptide according to SEQ ID No. 1 or SEQ ID No. 2 according to the invention, a nucleic acid according to the invention or an expression vector according to the invention, and a pharmaceutically acceptable carrier.
  • This composition is used for parenteral administration, such as subcutaneous, intradermal, intraperitoneal, intravenous, intramuscular or oral administration.
  • the peptides are dissolved or suspended in a pharmaceutically acceptable, preferably aqueous carrier.
  • the composition can contain excipients, such as buffers, binding agents, blasting agents, diluents, flavours, lubricants, etc..
  • the peptides can also be administered together with immune stimulating substances, such as cytokines.
  • immune stimulating substances such as cytokines.
  • An extensive listing of excipients that can be used in such a composition can be, for example, taken from A. Kibbe, Handbook of Pharmaceutical Excipients, 3. Ed., 2000, American Pharmaceutical Association and pharmaceutical press.
  • the composition can be used for a prevention, prophylaxis and/or therapy of adenomateous or cancerous diseases.
  • the pharmaceutical preparation containing at least one of the peptides of the present invention comprising SEQ ID No. 1 and/or SEQ ID No. 2, a nucleic acid according to the invention or an expression vector according to the invention, is administered to a patient that suffers from an adenomateous or cancerous disease that is associated with the respective peptide or antigen. By this, a T-cell-mediated immune response can be triggered.
  • the pharmaceutical composition according to the present invention preferably further comprises at least one additional tumour associated peptide comprising a sequence according to any of SEQ ID No. 3 to SEQ ID No. 10, a respective nucleic acid or a respective expression vector.
  • the peptides that are present in the pharmaceutical composition according to the invention have the same properties as described above for peptides of the present invention comprising SEQ ID No. 1 and/or SEQ ID No. 2.
  • they can have an overall length of between 9 and 100, preferably between 9 and 30, and most preferred between 9 and 16 amino acids.
  • at least one peptide according to any of SEQ ID No. 1 to SEQ ID No .11 can include non-peptide bonds.
  • the respective nucleic acids can encode for between 9 and 100, preferably between 9 and 30, and most preferred between 9 and 16 amino acids.
  • composition according to the invention that comprises (in particular tumour associated) peptides consisting of amino acid sequences according to SEQ ID No. 1 and/or SEQ ID No. 2 and SEQ ID No. 3 to SEQ ID No. 11.
  • compositions wherein the amount of (in particular tumour associated) peptide(s), of nucleic acid(s) according to the invention or expression vector(s) according to the invention as present in said composition is/are tissue, cancer, and/or patient-specific.
  • the peptide may also be tagged, or be a fusion protein, or be a hybrid molecule.
  • the peptide may be substantially pure, or combined with an immune-stimulating adjuvant, or used in combination with immune-stimulatory cytokines, or be administered with a suitable delivery system, for example liposomes.
  • adjuvants include Aquila's QS21 stimulon (Aquila Biotech, Worcester, MA, USA) which is derived from saponin, mycobacterial extracts and synthetic bacterial cell wall mimics, and proprietory adjuvants such as Ribi's Detox.
  • Quil A another saponin derived adjuvant, may also be used (Superfos, Denmark).
  • Other adjuvants such as Freund's may also be useful. It may also be useful to give the peptide conjugated to keyhole limpet hemocyanin (KLH) or mannan (see WO 95/18145 and Longenecker et al (1993) Ann. NY Acad. Sci. 690,276-291).
  • an adjuvans is defined as a substance enhancing the immune response to an antigen (MedlinePlus® Medical Dictionary, NIH)
  • other substances with this function may be used, including but not limited to toll-like receptor agonists (TLR agonists), preferably substances that interact agonistically with TLR 3, 7, 8, and 9, more preferably TLR 9, such as protamine-stabilising RNA, CpG- oligonucleotides, CpR-oligonucleotides, bacterial DNA, imidazoquinolines etc.
  • iNOS inducible nitric oxide synthase
  • ARGl arginase
  • IDO indoleamine-2,3-dioxygenase
  • VEGFR- 1 vascular endothelial growth factor receptor 1
  • VEGF vascular endothelial growth factor
  • COX-2 cyclooxygenase-2
  • TGF-beta receptor I TGF-beta-RI
  • inhibitors may be, for example, monoclonal antibodies against said molecules or small molecules.
  • Small molecules and monoclonal antibodies known in the art to have an inhibitory function towords the factors mentioned above, and thus an immune response enhancing effect are, for example, 1-MT, NCX-4016, rofecoxib, celebrex, BEC, ABH, nor-NOHA, SB-505124, SD-208, LY580276, AMD3100, axitinib, bevacizumab, JSI- 124, CPA-7, XL-999, ZD2171, pazopanib, CP-547632, and VEGF Trap.
  • substances reducing the number of regulatory T-cells are suitable as an adjuvans.
  • cyclophosphamide Cytoxan
  • ONTAK denileukin diftitox
  • Sunitinib anti-CTLA-4 (MDX-OlO, CP-675206), anti-CD25, anti-CCL22, and anti-GITR.
  • the vaccine is a nucleic acid vaccine. It is known that inoculation with a nucleic acid vaccine, such as a DNA vaccine, encoding a polypeptide leads to a T-cell response. It may be administered directly into the patient, into the affected organ or systemically, or applied ex vivo to cells derived from the patient or a human cell line which are subsequently administered to the patient, or used in vitro to select a subpopulation from immune cells derived from the patient, which are then re-administered to the patient.
  • a nucleic acid vaccine such as a DNA vaccine
  • nucleic acid is administered to cells in vitro, it may be useful for the cells to be transfected so as to co-express immune-stimulating cytokines, such as interleukin-2 or GM-CSF.
  • the nucleic acid vaccine may also be administered with an adjuvant such as BCG or alum. However, it is preferred if the nucleic acid vaccine is administered without adjuvant.
  • the polynucleotide may be substantially pure, or contained in a suitable vector or delivery system.
  • suitable vectors and delivery systems include viral, such as systems based on adenovirus, vaccinia virus, retroviruses, herpes virus, adeno-associated virus or hybrids containing elements of more than one virus.
  • Non- viral delivery systems include cationic lipids and cationic polymers as are well known in the art of DNA delivery.
  • Physical delivery such as via a "gene-gun" may also be used.
  • the peptide or peptide encoded by the nucleic acid may be a fusion protein, for example with an epitope from tetanus toxoid which stimulates CD4- positive T-cells.
  • any nucleic acid administered to the patient is sterile and pyrogen free.
  • Naked DNA may be given intramuscularly or intradermally or subcutaneously.
  • the peptides may be given intramuscularly, intradermally or subcutaneously.
  • the nucleic acid vaccine may comprise any suitable nucleic acid delivery means.
  • the nucleic acid preferably DNA, may be naked (i.e. with substantially no other components to be administered) or it may be delivered in a liposome or as part of a viral vector delivery system.
  • dendritic cells may be the mechanism of priming of the immune response; however, dendritic cells may not be transfected but are still important since they may pick up expressed peptide from transfected cells in the tissue ("cross-priming", e.g., Thomas AM, Santarsiero LM, Lutz ER, Armstrong TD, Chen YC, Huang LQ, Laheru DA, Goggins M, Hruban RH, Jaffee EM.
  • cross-priming e.g., Thomas AM, Santarsiero LM, Lutz ER, Armstrong TD, Chen YC, Huang LQ, Laheru DA, Goggins M, Hruban RH, Jaffee EM.
  • Cross-priming e.g., Thomas AM, Santarsiero LM, Lutz ER, Armstrong TD, Chen YC, Huang LQ, Laheru DA, Goggins M, Hruban RH, Jaffee EM.
  • nucleic acid vaccine such as DNA vaccine
  • nucleic acid vaccine is administered into the muscle
  • peptide vaccines are preferably administered s.c. or i.d.
  • vaccine is administered into the skin.
  • the nucleic acid vaccine may be administered without adjuvant.
  • the nucleic acid vaccine may also be administered with an adjuvant such as BCG or alum.
  • suitable adjuvants include Aquila's QS21 stimulon (Aquila Biotech, Worcester, MA, USA) which is derived from saponin, mycobacterial extracts and synthetic bacterial cell wall mimics, and proprietory adjuvants such as Ribi's Detox.
  • Quil A another saponin derived adjuvant, may also be used (Superfos, Denmark). It is preferred if the nucleic acid vaccine is administered without adjuvant. Other adjuvants such as Freund's may also be useful. It may also be useful to give the peptide conjugated to keyhole limpet haemocyanin, preferably also with an adjuvant.
  • targeting vectors may comprise a tissue-or tumour-specific promoter which directs expression of the antigen at a suitable place.
  • the invention in a further aspect thereof relates to a pharmaceutical composition, that contains one or more of said peptides according to the invention.
  • This composition is used for parenteral administration, such as subcutaneous, intradermal, intramuscular or oral administration.
  • the peptides are dissolved or suspended in a pharmaceutically acceptable, preferably aqueous carrier.
  • the composition can contain excipients, such as buffers, binding agents, blasting agents, diluents, flavours, lubricants, etc..
  • the peptides can also be administered together with immune stimulating substances, such as cytokines.
  • An extensive listing of excipients that can be used in such a composition can be, for example, taken from A. Kibbe, Handbook of Pharmaceutical Excipients, 3. Ed., 2000, American Pharmaceutical Association and pharmaceutical press.
  • the composition can be used for a prevention, prophylaxis and/or therapy of adenomateous or cancerous diseases.
  • the pharmaceutical preparation containing at least one of the peptides of the present invention comprising SEQ ID No. 1 and/or SEQ ID No. 2 is administered to a patient that suffers from a adenomateous or cancerous disease that is associated with the respective peptide or antigen.
  • a CTL-specific immune response can be triggered.
  • a combination of two or several peptides according to the present invention can be used as vaccine, either in direct combination or within the same treatment regimen.
  • combinations with other peptides for example MHC class I or II specific peptides can be used.
  • the person of skill will be able to select preferred combinations of immunogenic peptides by testing, for example, the generation of T-cells in vitro as well as their efficiency and overall presence, the proliferation, affinity and expansion of certain T-cells for certain peptides, and the functionality of the T-cells, e.g. by analysing the IFN- ⁇ production (see also examples below). Usually, the most efficient peptides are then combined as a vaccine for the purposes as described above.
  • a suitable vaccine will preferably contain between 1 and 20 peptides, more preferably 2, 3, 4, 5, 6, 7, 8, 9, 10 or 11 different peptides, further preferred 6, 7, 8, 9, 10 or 11 different peptides, and most preferably 11 different peptides.
  • the length of the peptide for use in a cancer vaccine may be any suitable peptide. In particular, it may be a suitable 9-mer peptide or a suitable 7-mer or 8-mer or 10-mer or 11-mer peptide or 12-mer. Longer peptides may also be suitable, 9-mer or 10-mer peptides as described in the attached Table 1 are preferred for MHC class I-peptides.
  • the peptide(s) constitute(s) a tumour or cancer vaccine. It may be administered directly into the patient, into the affected organ or systemically, or applied ex vivo to cells derived from the patient or a human cell line which are subsequently administered to the patient, or used in vitro to select a subpopulation from immune cells derived from the patient, which are then re- administered to the patient.
  • the peptide may also be conjugated to a suitable carrier such as keyhole limpet haemocyanin (KLH) or mannan (see WO 95/18145 and Longenecker et al (1993) Ann. NY Acad. Sci. 690,276-291).
  • KLH keyhole limpet haemocyanin
  • mannan see WO 95/18145 and Longenecker et al (1993) Ann. NY Acad. Sci. 690,276-291.
  • the peptide vaccine may be administered without adjuvant.
  • the peptide vaccine may also be administered with an adjuvant such as BCG or alum.
  • adjuvants include Aquila's QS21 stimulon (Aquila Biotech, Worcester, MA, USA) which is derived from saponin, mycobacterial extracts and synthetic bacterial cell wall mimics, and proprietory adjuvants such as Ribi's Detox.
  • Quil A another saponin derived adjuvant, may also be used (Superfos, Denmark).
  • Other adjuvants such as Freund's may also be useful. It may also be useful to give the peptide conjugated to keyhole limpet haemocyanin, preferably also with an adjuvant.
  • Other adjuvants such as those mentioned above, may be used.
  • the peptide may also be tagged, or be a fusion protein, or be a hybrid molecule.
  • the peptides whose sequence is given in the present invention are expected to stimulate CD8 + CTL. However, stimulation is more efficient in the presence of help provided by CD4 + T-cells.
  • the fusion partner or sections of a hybrid molecule suitably provide epitopes which stimulate CD4 + T-cells.
  • CD4 + stimulating epitopes are well known in the art and include those identified in tetanus toxoid.
  • said vaccine is a multiple peptide tumour vaccine for treatment of renal cell carcinoma.
  • said vaccine comprises a set of tumour-associated peptides according to SEQ ID No. 1 to 10 which are located and have been identified on primary renal cancer cells.
  • This set includes HLA class I and class II peptides.
  • the peptide set can also contain at least one peptide, such as from HBV core antigen, used as a positive control peptide serving as immune marker to test the efficiency of the intradermal administration.
  • the vaccine consists of 11 individual peptides (according to SEQ ID No.
  • peptides 1 to 11 with between about 1500 ⁇ g to about 75 ⁇ g, preferably between about 1000 ⁇ g to about 750 ⁇ g and more preferred between about 500 ⁇ g to about 600 ⁇ g, and most preferred about 578 ⁇ g of each peptide, all of which may be purified by HPLC and ion exchange chromatography and appear as a white to off-white powder.
  • the lyophilisate is preferably dissolved in sodium hydrogen carbonate, and is used for intradermal injection within 30 min after reconstitution at room temperature.
  • preferred amounts of peptides can vary between about 0.1 and 100 mg, preferably between about 0.1 to 1 mg, and most preferred between about 300 ⁇ g to 800 ⁇ g per 500 ⁇ l of solution.
  • the term "about” shall mean +/- 10 percent of the given value, if not stated differently.
  • the person of skill will be able to adjust the actual amount of peptide to be used based on several factors, such as, for example, the immune status of the individual patient and/or the amount of TUMAP that is presented in a particular type of cancer.
  • the peptides of the present invention might be provided in other suitable forms (sterile solutions, etc.) instead of a lyophilisate.
  • CD8-positive T-cells Some of the peptides whose sequence is given in the present invention are expected to stimulate CD8-positive T-cells (CTL). However, stimulation is more efficient in the presence of help provided by CD4-positive T-cells.
  • the fusion partner or sections of a hybrid molecule suitably provide epitopes which stimulate CD4-positive T-cells.
  • CD4-positive stimulating epitopes are well known in the art and include those identified in tetanus toxoid or the peptide from MMP7 provided by this invention.
  • the vaccine according to the invention can be dependent from the specific type of cancer that the patient to be treated is suffering from as well as the status of the disease, earlier treatment regimens, the immune status of the patient, and, of course, the HLA- haplotype of the patient.
  • the vaccine according to the invention can contain individualised components, according to personal needs of the particular patient. Examples are different amounts of peptides according to the expression of the related TAAs in said particular patient, unwanted side-effects due to personal allergies or other treatments, and adjustments for secondary treatments following a first round or scheme of treatment.
  • a still further aspect of the present invention relates to the use of a peptide according to the invention, or of a polynucleotide or expression vector encoding such a peptide, in the manufacture of a medicament for killing target cells in a patient which target cells aberrantly express a polypeptide comprising an amino acid sequence of the invention.
  • Preferred is the use as a pharmaceutical composition that is an anti-cancer vaccine.
  • a still further aspect of the present invention provides the use of a peptide according to the invention, or of a polynucleotide or expression vector encoding such a peptide, for the manufacture of a medicament for inducing an immune response, in particular a cellular immune response, more particularly a T-cell mediated immune response against cells of solid tumours which cells express a human class I or II MHC molecule on their surface and present a polypeptide comprising an amino acid sequence of the invention.
  • tumour cells of solid tumours in contrast to healthy cells of the same tissue, express human HLA class II molecule on their surface. This fact has been described only once in Brasanac et al (Brasanac D, Markovic-Lipkovski J, Hadzi-Djokic J, Muller GA, Muller CA. Immunohistochemical analysis of HLA class II antigens and tumor infiltrating mononuclear cells in renal cell carcinoma: correlation with clinical and histopathological data. Neoplasma.
  • MoAb monoclonal antibodies
  • TIM tumour infiltrating mononuclear cells
  • a still further aspect of the present invention provides the use of a peptide according to the invention, or of a polynucleotide or expression vector encoding such a peptide, in the manufacture of a medicament for killing target cells in a patient whose target cells aberrantly express a polypeptide comprising an amino acid sequence as given in any of SEQ ID No. 1 to 10.
  • a further aspect of the invention thus provides methods for producing activated T lymphocytes in vivo or in vitro, whereby a first method comprises contacting in vitro T-cells with antigen-loaded human class I or II MHC molecules expressed on the surface of a suitable antigen-presenting cell for a period of time sufficient to activate, in an antigen specific manner, said T-cell wherein the antigen is a peptide according to the invention.
  • a second method which is more preferred, is described by Walter et al. (Walter S, Herrgen L, Schoor O, Jung G, Wernet D, Buhring HJ, Rammensee HG, Stevanovic S. Cutting edge: predetermined avidity of human CD8 T-cells expanded on calibrated MHC/anti-CD28-coated microspheres. J Immunol. 2003 Nov 15;171(10):4974-8).
  • the MHC class II molecules may be expressed on the surface of any suitable cell and it is preferred if the cell is one which does not naturally express MHC class II molecules (in which case the cell is transfected to express such a molecule) or, if it does, it is defective in the antigen-processing or antigen-presenting pathways. In this way, it is possible for the cell expressing the MHC class II molecule to be primed substantially completely with a chosen peptide antigen before activating the CTL.
  • the antigen-presenting cell typically has an MHC class I or II molecule on its surface and preferably is substantially incapable of itself loading said MHC class I or II molecule with the selected antigen. As is described in more detail below, the MHC class I or II molecule may readily be loaded with the selected antigen in vitro.
  • the mammalian cell lacks or has a reduced level or has reduced function of the TAP peptide transporter.
  • Suitable cells which lack the TAP peptide transporter include T2, RMA-S and Drosophila cells.
  • TAP is the Transporter Associated with antigen Processing.
  • the human peptide loading deficient cell line T2 is available from the American Type Culture Collection, 12301 Parklawn Drive, Rockville, Maryland 20852, USA under Catalogue No CRL 1992; the Drosophila cell line Schneider line 2 is available from the ATCC under Catalogue No CRL 19863; the mouse RMA-S cell line is described in Karre and Ljunggren (1985) J. Exp. Med. 162,1745.
  • said host cell before transfection expresses substantially no MHC class I molecules. It is also preferred if the stimulator cell expresses a molecule important for T-cell costimulation such as any of B7.1, B7.2, ICAM-I and LFA 3.
  • combinations of HLA molecules may also be used.
  • Allogeneic cells may also be used in the preparation of CTL and this method is described in detail in WO 97/26328, incorporated herein by reference.
  • other cells may be used to present antigens such as CHO cells, baculovirus-infected insects cells, bacteria, yeast, vaccinia-infected target cells.
  • plant viruses may be used (see, for example, Porta et al (1994) Virology 202, 449-955 which describes the development of cowpea mosaic virus as a high-yielding system for the presentation of foreign peptides.
  • the antigen-presenting cell comprises an expression vector as above.
  • the activated T-cells which are directed against the peptides of the invention are useful in therapy.
  • a further aspect of the invention provides activated T-cells obtainable by the foregoing methods of the invention.
  • a still further aspect of the invention provides activated T-cells which selectively recognise a cell which aberrantly expresses a polypeptide comprising an amino acid sequence of the invention.
  • the T-cells recognises the said cell by interacting with the HLA/peptide-complex (for example, binding).
  • the T-cells are useful in a method of killing target cells in a patient which target cells aberrantly express a polypeptide comprising an amino acid sequence of the invention wherein the patient is administered an effective number of the activated T-cells.
  • the T-cells which are administered to the patient may be derived from the patient and activated as described above (i.e. they are autologous T-cells). Alternatively, the T-cells are not from the patient but are from another individual.
  • the individual is a healthy individual.
  • healthy individual the inventors mean that the individual is generally in good health, preferably has a competent immune system and, more preferably, is not suffering from any disease which can be readily tested for, and detected.
  • the activated T-cells express a T-cell receptor (TCR) which is involved in recognising cells which express the aberrant polypeptide. It is useful if the cDNA encoding the TCR is cloned from the activated T-cells and transferred into a further T-cells for expression.
  • TCR T-cell receptor
  • the target cells for the CD4-positive T-cells can be cells of the tumour (which sometimes express MHC class II) and/or stromal cells surrounding the tumour (tumour cells) (which sometimes also express MHC class II).
  • the TCRs of T-cell clones of the invention specific for the peptides of the invention are cloned.
  • the TCR usage in the T-cells clones is determined using (i) TCR variable region- specific monoclonal antibodies and (ii) RT PCR with primers specific for Va and Vp gene families.
  • a cDNA library is prepared from poly-A mRNA extracted from the T-cells clones. Primers specific for the C-terminal portion of the TCR a and P chains and for the N-terminal portion of the identified Va and P segments are used.
  • the complete cDNA for the TCR a and P chain is amplified with a high fidelity DNA polymerase and the amplified products cloned into a suitable cloning vector.
  • the cloned a and P chain genes may be assembled into a single chain TCR by the method as described by Chung et al (1994) Proc. Natl. Acad. Sci. USA 91, 12654-12658. In this single chain construct the VaJ segment is followed by the V DJ segment, followed by the Cp segment followed by the transmembrane and cytoplasmic segment of the CD3 chain.
  • This single chain TCR is then inserted into a retroviral expression vector (a panel of vectors may be used based on their ability to infect mature human CD8- positive T lymphocytes and to mediate gene expression: the retroviral vector system Kat is one preferred possibility (see Finer et al (1994) Blood 83, 43).
  • High titre amphotrophic retrovirus are used to infect purified CD8-positive or CD4-positive T lymphocytes isolated from the peripheral blood of tumour patients (following a protocol published by Roberts et al (1994) Blood 84, 2878-2889, incorporated herein by reference).
  • Anti-CD3 antibodies are used to trigger proliferation of purified CD8-positive T-cells, which facilitates retroviral integration and stable expression of single chain TCRs.
  • the efficiency of retroviral transduction is determined by staining of infected CD8-positive T-cells with antibodies specific for the single chain TCR.
  • In vitro analysis of transduced CD8-positive T-cells establishes that they display the same tumour-specific killing as seen with the allo-restricted T-cells clone from which the TCR chains were originally cloned.
  • Populations of transduced CD8-positive T-cells with the expected specificity may be used for adoptive immunotherapy of the tumour patients. Patients may be treated with in between 10 8 to 10 11 autologous, transduced T-cells. Analogously to CD8-positive, transduced CD4-positive T helper cells carrying related constructs can be generated.
  • a further aspect of the invention provides a TCR which recognises a cell which aberrantly expresses a polypeptide comprising an amino acid sequence of the invention, the TCR being obtainable from the activated T-cells.
  • TCR functionally equivalent molecules to the TCR are included in the invention. These include any molecule which is functionally equivalent to a TCR which can perform the same function as a TCR.
  • TCR functionally equivalent molecules to the TCR.
  • these molecules include genetically engineered three-domain single-chain TCRs as made by the method described by Chung et al
  • the invention also includes a polynucleotide encoding the TCR or functionally equivalent molecule, and an expression vector encoding the TCR or functionally equivalent molecule thereof.
  • Expression vectors which are suitable for expressing the TCR of the invention include those described above in respect of expression of the peptides of the invention. It is, however, preferred that the expression vectors are ones that are able to express the TCR in a T-cells following transfection.
  • a further aspect of the invention provides a method of killing target cells in a patient which target cells aberrantly express a polypeptide comprising an amino acid sequence of the invention, the method comprising administering to the patient an effective amount of a peptide according to the invention, or an effective amount of a polynucleotide or an expression vector encoding a said peptide, or an effective number of T lymphocytes as defined above, wherein the amount of said peptide or amount of said polynucleotide or expression vector or T-cells is effective to provoke an anti-target cell immune response in said patient.
  • the target cell is typically a tumour or cancer cell, in particular cells of solid tumors that express a human MHC class I or II molecule on their surface and present a polypeptide comprising an amino acid sequence as given above.
  • a still further aspect of the invention provides a method of killing target cells in a patient whose target cells aberrantly express a polypeptide comprising an amino acid sequence of the invention, the method comprising the steps of (1) obtaining T-cells from the patient; (2) introducing into said cells a polynucleotide encoding a TCR, or a functionally equivalent molecule, as defined above; and (3) introducing the cells produced in step (2) into the patient.
  • a still further aspect of the invention provides a method of killing target cells in a patient whose target cells aberrantly express a polypeptide comprising an amino acid sequence as defined above, the method comprising the steps of (1) obtaining antigen presenting cells, such as dendritic cells, from said patient; (2) contacting said antigen presenting cells with a peptide as defined in the first or second or third aspects of the invention, or with a polynucleotide encoding such a peptide, ex vivo; and (3) reintroducing the so treated antigen presenting cells into the patient.
  • antigen presenting cells such as dendritic cells
  • the antigen presenting cells are dendritic cells.
  • the dendritic cells are autologous dendritic cells which are pulsed with an antigenic peptide.
  • the antigenic peptide may be any suitable antigenic peptide which gives rise to an appropriate T-cell response.
  • T- cell therapy using autologous dendritic cells pulsed with peptides from a tumour associated antigen is disclosed in Murphy et al (1996) The Prostate 29, 371-380 and Tjua et al (1997) The Prostate 32, 272-278.
  • the antigen presenting cells such as dendritic cells, are contacted with a polynucleotide which encodes a peptide of the invention.
  • the polynucleotide may be any suitable polynucleotide and it is preferred that it is capable of transducing the dendritic cell thus resulting in the presentation of a peptide and induction of immunity.
  • the polynucleotide may be comprised in a viral polynucleotide or virus.
  • adenovirus-transduced dendritic cells have been shown to induce antigen-specific antitumour immunity in relation to MUCl (see Gong et al (1997) Gene Ther. 4,1023-1028).
  • adenovirus-based systems may be used (see, for example, Wan et al (1997) Hum. Gene Ther. 8, 1355-1363); retroviral systems may be used (Specht et al (1997) J Exp. Med.
  • RNA may also be used (Ashley et al (1997) J. Exp. Med. 186, 1177 1182).
  • the target cells are cancer cells, more preferably renal or colon cancer cells.
  • HLA haplotype of the patient is determined prior to treatment.
  • HLA haplotyping may be carried out using any suitable method; such methods are well known in the art.
  • the invention includes in particular the use of the peptides of the invention (or polynucleotides encoding them) for active in vivo vaccination; for manipulation of autologous dendritic cells in vitro followed by introduction of the so-manipulated dendritic cells in vivo to activate T-cell responses; to activate autologous T-cells in vitro followed by adoptive therapy (i.e. the so-manipulated T-cells are introduced into the patient); and to activate T-cells from healthy donors (MHC matched or mismatched) ill vitro followed by adoptive therapy.
  • the vaccines of the present invention are administered to a host either alone or in combination with another cancer therapy to inhibit or suppress the formation of tumours.
  • the peptide vaccine may be administered without adjuvant.
  • the peptide vaccine may also be administered with an adjuvant such as BCG or alum. Other suitable adjuvants are also described above..
  • the peptides according to the invention can also be used as diagnostic reagents. Using the peptides it can be analysed, whether in a T-cell-population T-cells are present that are specifically directed against a peptide or are induced by a therapy. Furthermore, the increase of precursor T-cells can be tested with those peptides that display reactivity against the defined peptide. Furthermore, the peptide can be used as marker in order to monitor the progression of the disease of a tumour that expresses said antigen of which the peptide is derived from.
  • the peptides are used for staining of leukocytes, in particular of T-lymphocytes. This use is of particular advantage if it should be proven, whether in a CTL-population specific CTLs are present that are directed against a peptide. Furthermore the peptide can be used as marker for determining the progression of a therapy in an adenomateous or cancerous disease or disorder.
  • the peptides are used for the production of an antibody.
  • Polyclonal antibodies can be obtained in a standard fashion by Immunisation of Animals via injection of the peptide and subsequent purification of the immune globulin.
  • Monoclonal antibodies can be produced according to standard protocols such as described, for example, in Methods Enzymol. (1986), 121, Hybridoma technology and monoclonal antibodies.
  • helper T-cell epitopes of TAA remains an important task in anti-tumour immunotherapy.
  • different strategies for the identification of class I or II peptides from TAA have been carried out, ranging from the incubation of APCs with the antigen of interest in order to be taken up and processed (Chaux, P., V. Vantomme, V. Stroobant, K. Thielemans, J. Corthals, R. Luiten, A.M. Eggermont, T. Boon, and B.P. van der Bruggen. 1999. Identification of MAGE-3 epitopes presented by HLA-DR molecules to CD4(+) T lymphocytes. J. Exp. Med.
  • the inventors identified a ligand accounting for one core sequence from MMP7.
  • the inventors found this protein to be over-expressed in renal cell carcinomas, in addition, it has been described as tumour-associated (Miyamoto, S., K. Yano, S. Sugimoto, G. Ishii, T. Hasebe, Y. Endoh, K. Kodama, M. Goya, T. Chiba, and A. Ochiai. 2004.
  • Matrix metalloproteinase-7 facilitates insulin-like growth factor bioavailability through its proteinase activity on insulin-like growth factor binding protein 3. Cancer Res. 64:665-671 ; Sumi, T., T. Nakatani, H. Yoshida, Y. Hyun, T.
  • SEQ ID No 1 to SEQ ID No 2 show peptide sequences of T-cell epitope containing peptides that are presented by MHC class I or II according to the present invention.
  • SEQ ID No 3 to SEQ ID No 11 show peptide sequences of peptides that are used in the vaccine of the present invention,which is subsequently referred to as "IMA".
  • Figure 1 shows the presentation of the c-Met protooncogene derived peptide IMA-MET-OOl on primary tumour sample RCCOl 3.
  • Nanocapillary high-performance liquid chromatography ESI MS was done on peptides eluted from RCCOl 3.
  • the mass chromatogram for 1006.54 ⁇ 0.5 Da shows a peak at retention time 47.8 min.
  • Collisionally induced decay mass spectrum from m/z 1006.54 recorded in a second LC-MS run at the given retention time and shown in the inset, confirmed the presence of IMA-MET-001 (Weinschenk 2002).
  • Figure 2 shows the tissue expression of c-Met protooncogene (MET). Expression was analyzed by oligonucleotide microarrays. Copy numbers are relative to kidney, which is set at 1.0. "P” means that the gene is present, “A” absent and “M” marginal according to the statistical absolute call algorithms. “I” means that expression of the gene is significantly increased relative to kidney, “D” stands for decreased expression, and “NC” means that there is no change in expression. The expression value relative to kidney is calculated from the signal log ratio and displayed on top of the bars. The dashed horizontal line shows the highest expression in normal tissues (in this case lung).
  • MET c-Met protooncogene
  • Figure 3 shows the killing of peptide-loaded target cells by CTLs primed with IMA-MET-001
  • Figure 4 shows the killing of malignant cells by CTL primed with IMA-MET-001.
  • Figure 5 shows the cold target inhibition assay.
  • Figure 6 shows the tetrameric analysis of microsphere driven expansions.
  • Figure 7 shows the in vitro immunogenicity of IMA-MMP-001 - Representative intracellular IFN gamma versus CD4 stainings of four healthy donors.
  • Donor 1 , 2 and 3 showed CD4- positive T-cells reactive against IMA-MMP-001 after the third and the fourth stimulation.
  • Donor 4 was always negative.
  • Figure 8 shows differential peptide presentation on tumour and healthy tissue -
  • A Mass spectrum of two peptide species m/z 739.96 and 741.95 derived from normal kidney and renal cell carcinoma tissue of patient RCClOO, respectively. The mass spectrum demonstrates an about 4-fold overpresentation of the Adipophilin peptide on the renal cell carcinoma tissue compared to the corresponding autologous normal tissue
  • B The collisionally induced decay mass spectrometry analysis of m/z 741.95 (tumour) revealed the peptide sequence IMA-ADF- 003, a peptide sequence derived from Adipophilin.
  • Figure 9 shows in vivo immunity against IMA-ADF-001 - T-cell immunity in 2 RCC patients against several non-vaccinated peptides in patients vaccinated with autologous dendritic cells pulsed with two TUMAPs derived from MUC.
  • T-cells specific for IMA-ADF-001 (“Adipophilin") were not present prior to vaccination and were detected in patient #3 (upper panel) after 6 vaccinations and in patient #8 (lower panel) after 8 vaccinations.
  • Figure 10 shows representative examples of an IMA induced T-cell response identified by the amplified ELISPOT assay for the same patient and antigen.
  • Upper and lower column represent negative control antigen HIV-001 and single TUMAP IMA-CCN-001 used for readout, respectively.
  • the left column shows ELISPOTs of pooled samples taken before vaccination on screening day 2 (S2) and immediately prior to the first injection (Vl).
  • the right column shows ELISPOTs of pooled samples taken during the vaccination protocol on day 22 (V6) and day 36 (V7). The number of positive cells is given for each experiment.
  • Figure 11 Representative examples of IMA induced T-cell responses identified by the amplified Tetramer staining assay.
  • Upper and middle panels represent two-dimensional dot plots gated on CD3+ lymphocytes, lower panels are gated on CD3+ CD8+ lymphocytes. Patients, timepoints and stainings as indicated for each column.
  • Sl +Vl samples taken prior to vaccination
  • V4+V5 samples taken at day 8 and day 15
  • V6+V7 samples taken on day 22 and day 36
  • V8+FU samples taken on day 64 (last vaccination) and after 85 to 92 days (end of study)
  • a cell population positive for CD3+ and IMA-CCN-001 tetramer can be identified after the forth and fifth injection of IMA (V6+V7; middle panel) accounting for 0.78% of the lymphocytes (V6+V7;lower panel). No positive population was found for the K67-001 tetramer (upper panel).
  • Figure 12 Observed T-cell magnitude kinetics in single time point amplified tetramer assays. Results are shown for single time point readouts for patient 05-001 for whom the vaccine- induced T-cell response had been detected by the routine amplified tetramer assay with pooled samples. Results are given for all tumour associated antigens present in IMA (TUMAP pool) and in particular for the IMA-CCN-001 peptide. Additionally, the HIV-001 and IMA- HBV-001 controls within the same single time point assay are shown.
  • GM-CSF rhuGM-CSF (recombinant human Granulocyte-Macrophage rhuGM-CSF Colony- Stimulating Factor)
  • Peptides that were identified from primary RCC tissue were selected for inclusion into the vaccine IMA (see below) according to an internal ranking system mainly based on gene expression analysis, literature, and database search for known properties of an antigen from which a derived peptide has been identified. All naturally presented peptides are highly over- expressed in renal cell carcinoma tissue compared to normal kidney tissue as well as a range of other vital organs and tissues. Such a selection is necessary (1) to select for peptides that are able to induce T-cells with high specificity for recognition of the tumour but not other tissue to minimize the chance of autoimmunity induced by the vaccination of IMA and (2) to ensure that the majority of tumours in a patient population is recognized by the induced T- cell.
  • RNA sources - Total RNA from human tissues were obtained commercially (Ambion, Huntingdon, UK; Clontech, Heidelberg, Germany; Stratagene, Amsterdam, The Netherlands, BioChain, Heidelberg, Germany). Total RNA from several individuals was mixed in a way that RNA from each individual was equally weighted. Quality and quantity was confirmed on the Agilent 2100 Bioanalyzer (Agilent, Waldbronn, Germany) using the RNA 6000 Nano LabChip Kit (Agilent).
  • Pairwise comparisons were calculated using the expression values in kidney as baseline. Accordingly, all expression values calculated from signal log ratios are relative to kidney, which was set at 1. Significance of differential expression was judged by the "change" values given by the statistical algorithms implemented in the software. For absolute detection of expression, data were analyzed again using the statistical algorithms. Presence or absence of gene expression was determined by the absolute call algorithms.
  • MET c-Met protooncogene
  • Table 1 summarizes the peptides contained in the vaccine of the invention IMA, comprising also the peptides according to the invention.
  • Table 2 summarizes the expression results for all antigens coding for peptides contained in the vaccine of the invention IMA, as well as for the peptides according to the invention.
  • MUC mesenchymal cells
  • Aberrant deglycosylation in malignancies is common and unmasks epitopes in tumour cells which might not be presented on normal cells. It is highly likely that such a mechanism also occurs in RCC. This would explain the specific killing of tumor cell lines expressing MUC (Brossart 1999). Please also see chapter 4.1.5 on the properties of MUC.
  • IMA-MUC-001 has been administered in conjunction with autologous dendritic cells in an investigator-initiated trial at the University of Tubingen. In this trial presented recently at the ASCO 2003 (Mueller 2003) and follow-up data at the ASCO 2005 (Wierecky 2005) Meeting no autoimmune effects were reported.
  • IMA-MUC-001 can be considered as safe although no over-expression can be detected for the MUC antigen on mRNA level alone.
  • IMA-MMP-001 is a peptide binding to HLA-DR, a HLA class II molecule.
  • Class II TUMAPs activate T helper cells which play a crucial role in assisting the function of cytotoxic T-cells activated by class II TUMAPs.
  • Promiscuous binding of a HLA-DR peptide is important to ensure that the majority (>50%) of the HLA-A* 02-positive patients treated with IMA are also able to elicit a T-cell response to IMA-MMP-001.
  • IMA-MMP-001 binds promiscuously to several HLA-DR alleles (DRBl + OlOl, *0301, *0401, * 1101 and *1501) covering a total of at least 69.6% of the HLA- A2 positive Caucasian population. Promiscuous binding of IMA-MMP-001 is confirmed experimentally by in vitro immunogenicity data.
  • IMA-MMP-001 binding of IMA-MMP-001 to several common HLA-DR alleles (see table below) was ranked.
  • the algorithm has been successfully used to identify class I and class II epitopes from a wide range of antigens, e.g. from the human TAA TRP2 (class I) (Sun 2000) and SSX2 (class II) (Neumann 2004).
  • the analyzed HLA-DR alleles cover at least 69.6% of the HLA- A2 positive Caucasian population (Mori 1997).
  • the threshold for binding was defined at a score of 18 based on the analysis of binding scores of known published promiscuous HLA-DR ligands.
  • Promiscuous binding is defined as binding of a HLA-DR peptide to several HLA-DR alleles expressed in at least 50% of the Caucasian population.
  • HLA-A and HLA-DR are in linkage disequilibrium yielding combinations of HLA-A2 and specific HLA-DRs that are favoured in comparison to others (Table 3).
  • Ligands of certain MHC molecules carry chemical related amino acids in certain positions of their primary sequence which permits the definition of a peptide motif for every MHC allele (FaIk 1991).
  • SYFPEITHI uses motif matrices deduced from refined motifs exclusively based on natural ligand analysis by Edman degradation and tandem mass spectrometry (Schirle 2001). These matrices allow the exact prediction of peptides from a given protein sequence presented on MHC class I or class II molecules (Rotzschke 1991).
  • Table 4 Binding scores of IMA-MMP-001 to common HLA-DR alleles Shown are the IMA-MMP-001 SYFPEITHI binding scores for the most common HLA-DRBl alleles in the Caucasian population. The frequencies of the corresponding serological haplotypes of HLA- A2 positive Caucasians are given in brackets. The peptide was considered as binding to a HLA molecule when the score was equal or higher than 18.
  • IMA-MMP-001 is likely to bind to several HLA-DR alleles (DRBl + OlOl, *0301, *0401, *1101 and *1501) covering at least 69.6 % of the HLA- A2 positive Caucasian population. As no frequency data of HLA-DRl 5 is available this allele was omitted in the calculation. Thus, it is very likely that the coverage of the population is even higher than 69.9%. Experimental confirmation for promiscuous binding of IMA-MMP-001 is obtained by in vitro immunogenicity data (see below).
  • Overexpressed antigens are supposed to be overpresented on HLA molecules on the cell surface.
  • HLA-A*03 peptide derived from Adipophilin an overexpressed antigen from which the HLA-A*02 peptides IMA-ADF-001 and IMA-ADF-002, both contained in IMA, are derived, was shown to be highly overpresented on renal cell carcinoma tissue compared to the autologous normal tissue from patient RCClOO employing the QUALITEA strategy. This demonstrates in this exemplary case that overexpression of an antigen (in this case Adipophilin) correlates with overpresentation of derived peptides of the same antigen.
  • QUALITEA represents a strategy for differential quantitation of HLA-eluted peptides from tumour and normal tissue.
  • HLA ligands derived from the two different sources are N-terminally derivatised either by a 1 H x - or D x - reagent and combined.
  • peptides are quantitated by ESI-MS analysis according to their peak areas.
  • a pair of derivatised peptides ( ⁇ x -derivatisation and 2 D x -derivatisation) is physico- chemically identical and easily detectable because it essentially coelutes in chromatographic systems.
  • FIG. 8 An example for differential HLA peptide presentation on tumour and normal tissue of an overexpressed antigen is shown in Figure 8.
  • the peptide IMA-ADF-003 which was 4-fold overpresented on tumour tissue vs. healthy kidney tissue from patient RCClOO was identified by collisionally induced decay mass spectrometry analysis among many equally presented peptides.
  • This peptide overpresented on tumour tissue was derived from Adipophilin.
  • Gene expression analysis of the same patient RCClOO revealed also a 2.64 fold overexpression of Adipophilin in this tumour tissue compared to healthy kidney (data not shown). This data confirm in this particular case that overexpression in tumour tissue on gene level leads to peptide overpresentation on the tumour cell surface.
  • APCs antigen-presenting cells
  • TUMAP T-cell epitope
  • IMA is a vaccine containing a set of tumour-associated peptides which are located and have been identified on primary renal cancer cells. This set includes HLA class I and class II peptides. The peptide set also contains one peptide from HBV core antigen used as a positive control peptide serving as immune marker to test the efficiency of the intradermal administration. Peptide vaccination in general needs to be adjuvanted, and such, GM-CSF will be exploited as adjuvant in this vaccination schedule (Human GM-CSF is commercially available as Sargramostim, Leukine®, Berlex).
  • IMA-MUC-001 and IMA-CCN-001 were identified using other technologies. For both latter peptides, natural presentation of these peptides by tumour cell lines is demonstrated based on indirect evidence in the in vitro immunogenicity assay (see below).
  • HLA molecules from shock-frozen processed primary renal cell carcinoma tissue are purified and HLA-associated peptides are isolated. These peptides either are separated off-line by HPLC and fractions are analyzed or sequence analysis by mass spectrometry is done by online HPLC-MSMS experiments. The resulting sequences are verified by synthesis of the identified peptides and comparison of the fragment spectra of identified and synthesized peptides. As the identified peptides are directly derived from HLA molecules of the primary tumours these results present direct evidence on the natural processing and presentation of the identified peptides on primary renal cell carcinoma tissue.
  • HLA molecules were purified by affinity chromatography using the HLA class I-specific antibody W6/32 or the HLA-A* 02-specific antibody BB7.2 or (in the case of IMA-MMP-001) the HLA-DR-specific antibody L243.
  • HLA-associated peptides were eluted by acid treatment and isolated from the MHC alpha chain-protein by ultrafiltration.
  • the isolated peptides either were separated off-line by reversed-phase high performance liquid chromatography and fractions were analyzed by nano-ESI MS on a hybrid quadrupole orthogonal acceleration time-of-flight tandem mass spectrometer (Q-TOF I or Q-TOF Ultima, Waters) or on-line LC-MSMS analysis was done using the same instruments. A blank run was always included to ensure that the system was free of peptide. Calibrations were done at least once per day and analyses on standard compounds were done in appropriate intervals to guarantee optimal performance of the systems. Interpretation of the fragment spectra was done manually.
  • Verification of the analysis was obtained by database searches and solid-phase synthesis of the putative peptide sequence and comparison of the fragment spectra of identified and synthesized peptide. All peptides contained in IMA (data not shown) except IMA-MUC-OOl and IMA-CCN-OOl were identified in the identical fashion confirming the natural presentation of these peptides on primary renal cell carcinoma tissue.
  • the drug product IMA is presented as a lyophilisate for intradermal application containing 1 1 peptides - 578 ⁇ g of each peptide - in form of their salts (acetates).
  • the powder for injection containing 578 ⁇ g of each peptide will be dissolved in 700 ⁇ l sodium hydrogen carbonate (4.2 %).
  • 500 ⁇ l (equals a single dose of 413 ⁇ g of each peptide per injection and a total single dose of 4.5 mg IMA per injection) will be injected intradermally.
  • IMA contains 9 HLA class I tumour-associated peptides, 1 HLA class II tumour-associated peptide and 1 HLA-class I viral control peptide. In vitro immunogenicity could be demonstrated for the vast majority of peptides contained in IMA.
  • Killer assay cytotoxic killing of target measured by chromium release assay; Cytokine release: release of cytokines by T-cells measured by ELISA; Cytokine staining: synthesis of cytokines by T-cells measured by intracellular flow cytometry; Tetramer detection: detection of peptide-specific T-cells by HLA tetramers.
  • IMA contains 10 HLA class I-binding peptides.
  • CD8-positive cytotoxic T-cells were generated from autologous peripheral blood mononuclear cells (PBMC) from healthy donors using single peptides contained in IMA and the activity of these cytotoxic T-cells was tested with chromium release assays and detection of T-cells with HLA tetramers in flow cytometry.
  • PBMC peripheral blood mononuclear cells
  • cytotoxic T-cells are generated (primed) in vitro by repeated stimulation of peripheral blood mononuclear cells (PBMC) from healthy HLA-A* 02 positive donors with the specific peptide to be tested.
  • PBMC peripheral blood mononuclear cells
  • the priming can be done either using autologous dendritic cells generated from blood monocytes of the donor or using HLA tetramer-loaded beads.
  • A. Cytotoxic killing of target In the second step, cytoxicity of such primed cytotoxic T-cells (CTLs) are tested by labelling target cells with radioactive chromium and incubating target cells with generated CTLs. The amount of radioactive chromium released into the supernatant can be correlated directly to proportion of killed target cells.
  • CTLs cytotoxic T-cells
  • B. Detection of T-cells with HLA tetramers Alternatively, in the second step primed CTLs with specificity for a given peptide are detected using HLA tetramers. Tetramers consist of four peptide-loaded HLA-A*02 molecules coupled to each other.
  • constructs allow specific labelling of the cognate T-cell receptor that recognizes the HLA-peptide-complex in the tetramer and by labelling of the tetramer with a fluorochrome followed by analysis in flow cytometry (FACS).
  • biotin-FITC/mg microspheres 0.06 ⁇ g biotin-FITC/mg microspheres (Bangs Laboratories, Fishers, Illinois/USA).
  • a sterile PBS/BSA/EDTA buffer was used for coupling to biotinylated molecules.
  • microspheres were washed and resuspended at 2 x 10 6 particles/ml in buffer containing biotinylated MHC and / or antibodies at various concentrations. Binding was allowed at room temperature for 30 min while agitating. Coated beads were washed three times, resuspended in above buffer and stored for up to 4 weeks at 4 0 C before use.
  • PBMCs peripheral blood mononuclear cells
  • PBMCs peripheral blood mononuclear cells
  • untouched CD8 T-cells were magnetically enriched by negative depletion using a CD8 T-cell isolation kit (Miltenyi Biotec, Bergisch Gladbach, Germany) according to the manufacturer's conditions, which resulted in a purity of CD8-positive TCR-positive cells of more than 80%.
  • In vitro stimulations were initiated in 24 well plates with 5 * 10 6 responder cells plus 1 x 10 6 APCs or microspheres per well in 1.5 ml T-cell medium.
  • the standard 51 Cr-release assay was performed as described (Brossart 2001). Target cells were pulsed with 50 ⁇ g/ml peptide for 2 h and labelled with 51 Cr-sodium chromate in RPlO for 1 h at 37°C. 10 4 cells were transferred to a well of a round-bottomed 96-well plate. Varying numbers of CTL were added to give a final volume of 200 ⁇ l and incubated for 4 h at 37° C. At the end of the assay supernatants (50 ⁇ l/well) were harvested and counted in a beta- plate counter. The percent specific lysis was calculated as: 100 x (experimental release - spontaneous release / maximal release-spontaneous release).
  • Tetramer staining was performed at indicated before (Walter 2003) or with minor modifications. Briefly, biotinylated recombinant HLA-A*0201 molecules lacking the transmembrane domain and being biotinylated at the carboxy terminus of the heavy chain were produced as previously described (Airman 1996). Fluorescent tetramers were generated by coincubating biotinylated HLA-A* 0201 with streptavidin-PE or streptavidin-APC (Molecular Probes, Leiden, The Netherlands) at a 4:1 molar ratio.
  • the cytotoxicity of the induced CTL was analyzed in a standard 51 Cr-release assay using peptide loaded T2 cells and autologous DC as targets.
  • the CTL line obtained after two weekly restimulations demonstrated antigen-specific killing.
  • the T-cells only recognized T2 cells or DC coated with the cognate peptide while they did not lyse target cells pulsed with irrelevant HLA-A2 binding peptides derived from survivin protein or HIV-I reverse transcriptase confirming the specificity of the cytolytic activity.
  • HLA-A*02 positive cell lines HCT 116 (colon cancer), A498, MZ 1257 (renal cell carcinoma, RCC), MCF-7 (breast cancer), Mel 1479 (malignant melanoma) and U266 (multiple myeloma) that express c-Met as targets in a standard 51 Cr-release assay.
  • the EBV-transformed B-cell line Croft (HLA-2+/c-Met-) and the ovarian cancer cell line SK-OV-3 (HLA-A3+/c-Met+) were included to determine the specificity and HLA-restriction of the CTL.
  • the c-Met peptide specific CTL were able to efficiently lyse malignant cells expressing both HLA-A2 and c- Met.
  • There was no recognition of the ovarian cancer cells SK-OV-3 or Croft cells demonstrating that the presentation of c-Met peptide in context of HLA- A2 molecules on the tumour cells is required for the efficient lysis of target cells and confirm the antigen specificity and MHC restriction of the CTL.
  • the in vitro induced T-cells did not recognize the K 562 cells indicating that the cytotoxic activity was not NK-cell mediated.
  • Enriched CD8 T-cells of one A* 02+ healthy donor were stimulated 3 times with beads coated in the presence of 10 nM CD28 Ab plus either 10 nM of an irrelevant A*02 complex (left panel) or A*02 refolded with indicated antigens (middle and right panel).
  • Indicated Antigens were peptides NLVPMVATV from CMV pp65 (Wills 1996), modified peptide ELAGIGILTV from Melan-A/M ART-I (Kawakami 1994) and peptide IMA-MET-OOl.
  • T-cell lines were surface stained with CD8-PerCP Ab, cognate tetramer-PE (Figure 6; left and middle panel) and irrelevant A*02/ILKEPVHGV tetramer-APC (right panel). Percentage of tetramer+ cells among CD8-positive lymphocytes is indicated in each plot.
  • IMA contains one HLA class II peptide from matrix metalloproteinase 7, IMA-MMP-001.
  • CD4-positive T-cells were generated from autologous peripheral blood mononuclear cells (PBMC) from healthy donors with different HLA genotypes using the IMA-MMP-001 peptide and the activity of these Helper T-cells tested with intracellular cytokine staining in flow cytometry.
  • PBMC peripheral blood mononuclear cells
  • CD4-positive T-cells were generated (primed) in vitro by repeated stimulation of peripheral blood mononuclear cells (PBMC) from healthy donors with the specific peptide to be tested in the presence of IL- 12.
  • the priming was performed using autologous dendritic cells generated from blood monocytes of the donors.
  • the activity of primed CD4-positive T-cells specific for the given peptide were tested by measurement of IFN ⁇ production by intracellular IFN ⁇ staining using a fluorescently labelled antibody. The analysis was done by flow cytometry (FACS).
  • DCs dendritic cells
  • Human DCs were prepared out of PBMCs from freshly drawn blood from healthy donors.
  • PBMCs were isolated using a Ficoll density gradient (Lymphocyte Separation Medium, PAA Laboratories GmbH, Pasching, Austria). The obtained cells were washed, resuspended in X- Vivo 15 medium supplemented with 50 U/ml penicillin, 50 ⁇ g/ml streptomycin and 2mM L- Glutamine (BioWhittaker, Verviers, Belgium) and plated at a density of 7 x 10 6 cells/ml.
  • adherent monocytes were cultured for 6 days in X-Vivo medium with 100 ng/ml GM-CSF and 40 ng/ml IL-4 (AL-ImmunoTools, Friesoythe, Germany).
  • immature DCs were activated with 10 ng/ml TNF- ⁇ (R&D Systems, Wiesbaden, Germany) and 20 ⁇ g/ml poly(IC) (Sigma Aldrich, Steinheim, Germany) for 3 days.
  • the differentiation state of DCs was examined by flow cytometry, mature DCs being predominantly CD 14-, CD40-positive, CD80-positive, CD83-positive, CD86-positive and HLA-DR+ (data not shown).
  • PBMCs were stimulated with 2 x 10 5 autologous DCs. After priming, restimulations were done with cryopreserved autologous PBMCs every 6 to 8 days. For stimulation, cells were pulsed with 5 ⁇ g/ml peptide for 90' at 37°C and irradiated (60 Gy; Gammacell 1000 Elite, Nordion International Inc, Ontario, Canada).
  • T-cell medium RPMI 1640 containing HEPES and L-glutamin (Gibco, Paisley, UK) supplemented with 10% heat- inactivated human serum (PAA, C ⁇ lbe, Germany), 50 U/ml penicillin, 50 ⁇ g/ml streptomycin and 20 ⁇ g/ml gentamycin (BioWhittaker) in the presence of 10 ng/ml IL- 12 (Promocell, Heidelberg, Germany).
  • PBMCs Cryopreserved PBMCs were thawed, washed two times in X-Vivo 15 medium, resuspended at 10 7 cells/ml in T-cell medium and cultured overnight to reduce unspecific IFN ⁇ production (Provenzano 2002).
  • PBMCs were pulsed with 5 ⁇ g/ml peptide for 2 h, washed three times with X-Vivo 15 medium and incubated with effector cells in a ratio of 1 : 1 for 6 h.
  • Golgi-Stop Becton Dickinson, Heidelberg, Germany
  • Cells were analyzed using a Cytofix/Cytoperm Plus kit (Becton Dickinson) and CD4-FITC- (Immunotools), IFN ⁇ -PE- and CD8-PerCP clone SKl -antibodies (Becton Dickinson). After staining, cells were analyzed on a three-color FACSCalibur (Becton Dickinson). To generate antigen-specific CD4-positive T-cells and to test the peptide on promiscuous binding, PBMCs of 4 healthy donors with different HLA-DR alleles ( Figure 7) were stimulated using peptide-pulsed autologous DCs.
  • IFN ⁇ production was assessed by flow cytometry.
  • T- cells were analyzed after the third and the fourth stimulation by intracellular IFN ⁇ staining plus CD4-FITC and CD8-PerCP staining to determine the percentage of IFN ⁇ -producing cells in specific T-cell subpopulations.
  • stimulations with irrelevant peptide and without peptide were performed as negative controls.
  • IFN ⁇ response was considered as positive if the detection of IFN ⁇ producing CD4-positive T-cells was more than two fold higher compared to negative control. (Horton 2004).
  • In three of four donors we were able to generate CD4-positive T-cells specifically reacting to the peptide of interest (Figure 7). T-cell responses could not be observed in Donor 4 neither after the third nor after the fourth stimulation.
  • the highest frequencies of IFN ⁇ producing CD4-positive T-cells specific for IMA-MMP-001 were seen in Donor 1 and 2, respectively.
  • IMA-MMP-001 is a promiscuous binder being able to elicit CD4-positive T-cell responses in three out of four healthy donors carrying different HLA alleles.
  • All four alleles have a Glycine residue at position 86 and an Aspartic acid residue at position 57 of their ⁇ chains (data not shown). Therefore, they have very similar binding motives sharing binding characteristics for their binding pockets Pl and P4 (Rammensee 1997; Hammer 1993; Marshall 1994).
  • Donor 4 carries with HLA-DRBl*0318 and DRB1*14O1 alleles with very different binding motifs. This would explain why it was not possible to elicit a T-cell response with cells from this donor using the peptides mentioned above.
  • immunomonitoring has been a common practice in thousands of patients in various therapeutic vaccination studies (Romero 2004). Many different immunomonitoring methods have been reported to date, including functional and specific assays.
  • the immunogenic components of the therapeutic vaccine IMA are HL A-binding peptides which are supposed to induce specific T lymphocytes in vivo. This activation will lead to their proliferation and acquisition of effector functions, which includes their ability to secrete cytokines upon antigen contact.
  • ELISPOT assays As a surrogate marker for T-cell activation the frequency of specific cytokine secreting mononuclear cells in blood can be tested using ELISPOT assays.
  • Such assays are especially appropriate for this purpose as they are single cell based and result in a parameter (i. e. the number of spot forming cells among mononuclear cells) that is expected to be directly correlated to the true frequency of cytokine secreting antigen specific T lymphocytes in blood samples.
  • assays also enable the processing of relatively large numbers of samples and peptides in parallel, they have widely been used already for immunomonitoring studies in a large number of clinical studies so far (Schstoff 2000).
  • T-cell response analysis The immunological activity/efficacy can be described by T-cell response analysis.
  • T-cell responses of up to 100 % are described by Disis et al., 1999, in patients with ovarian and breast cancer.
  • Other authors have published results regarding T-cell responses between 33% and 83% in patients with melanoma (Keilholz/Schadendorf, 2003; Slingluff et al., 2004).
  • Gaudernack/Gjertsen, 2003 report about an immune response of several CD4-positive and CD8-positive T-cell clones specific for the antigen used in this study.
  • MUCl peptide specific T-cell responses in vivo were detected in the PBMC of all patients with OR. These in vivo induced T-cells were able to recognize target cells pulsed with the cognate peptide or matched allogeneic tumour cells (A498) constitutively expressing MUCl in an antigen and HLA restricted manner after in vivo restimulation.
  • T-cell responses to antigens not used for vaccination like adipophilin, telomerase or OFA could be detected indicating that epitope spreading might occur.
  • Proliferative response to the PADRE peptide were detectable in 11/16 patients, in some patients already after the 2nd vaccination.
  • IMA The clinical trial formulation of IMA consists of:
  • ex vivo ELISPOT In the "ex vivo ELISPOT" assay, cells are thawed from different time points, the number of live cells is counted and samples are assayed by one-day incubation with different peptides or controls in triplicate wells.
  • the ex vivo ELISPOT assay delivers quantitative data in a much quicker fashion compared to the other assays below. Additionally, this is the only assay that allows measurement of the one HLA class II peptide (IMA-MMP-OOl) contained in IMA. However, this assay is of limited sensitivity and positive data was only expected in the case of very strong T-cell responses comparable to memory (recall) immune responses to viruses.
  • ELISPOT enzyme linked sandwich antibody method
  • This assay has often been used to detect T-cell response to vaccinated tumour antigens in various third- party clinical trials. The possibility of false-positive data due to "in vitro priming" of T-cells is excluded by using various controls in this assay. Compared to the Tetramer Assay below, this assay gives additional functional information, i.e. IFN- ⁇ cytokine release.
  • the amplified ELISPOT assay was performed for 28 patients in the study and all antigens present in IMA prior and during the vaccination protocol at different time points.
  • Figure 10 shows representative examples of an IMA induced T-cell response identified by the amplified ELISPOT assay for the same patient and antigen.
  • Upper and lower column represent negative control antigen HIV-OOl and single TUMAP IMA-CCN-OOl used for readout, respectively. The number of positive cells is given for each experiment.
  • the left column shows ELISPOTs of pooled samples taken before vaccination while the right column shows ELISPOTs of pooled samples taken during the vaccination protocol.
  • lymphocyte gates and CD3/CD8 gates were identical for all stainings of one patient within an assay. Tetramer positive populations were identified from CD 8+ T-cells by analyzing double tetramer dot plots with quadrants or gates. Definition of tetramer+ cells was identical for each staining condition for a given patient in an assay and and based on recognizable cell populations. The amplified Tetramer assay was performed for 28 patients in the study and all antigens present in IMA prior and during the vaccination protocol at different time points.
  • Figure 11 shows two representative examples of IMA induced T-cell responses identified by the amplified Tetramer staining assay. Upper and middle panels represent two-dimensional dot plots gated on CD3+ lymphocytes, lower panels are gated on CD3+ CD8+ lymphocytes. Patients, timepoints and stainings were as indicated for each column.
  • Figure 11 A the immunological response to IMA-CCN-OOl in patient 03-004, that was already shown by ELISPOT assay ( Figure 10) was confirmed by tetramer assay.
  • a cell population positive for CD3+ and IMA-CCN-OOl tetramer was identified after the forth and fifth injection of IMA (V6+V7; middle panel) while no positive population was found for the K67-001 tetramer (upper panel).
  • the IMA-CCN-001 positive cells increased from 0.03% prior vaccination to 0.78% of the lymphocytes (V6+V7; lower panel) after the first three injections.
  • the amount of these cells increased from 0.02% prior vaccination to 0.8 % of the lymphocytes (lower panel; column 3) after the first three injections and decreased to 0.31% after day 64 (lower panel; column 4; V8+FU).
  • the amplified tetramer assay was performed for single time points, i.e. blood samples were not pooled, allowing a more precise evaluation of the T-cell kinetics an example of which is depicted in Figure 12.
  • the observed T-cell magnitude kinetics in single time point amplified tetramer assays are shown for patient 05-001.
  • tumour associated antigens present in IMA were highest on day 22, 36 and 64 (V6, V7 and V8) while response to the IMA-CCN-001 peptide peaked earlier at day 22 (V6).
  • the HBV-001 positive control resulted in an even faster response that reached its maximum at day 15 (V5).
  • Results are summarised in Table 9 below. Shown are all evaluable results from ex vivo tetramer and patient / antigen matched amplified tetramer assays where a vaccine-induced response was detected in the amplified assay. For the "ex vivo” method, % Tetramer+ among total CD8+ T-cells is indicated. As for the "amplified, routine” evaluation method, subpopulations of CD8+ T-lymphocytes may be analyzed, a second re-evaluation of the routine data is shown ("amplified, quantitative") with calculations based on total CD8+ T- lymphocytes. The "amplification factor" was calculated if a discrete tetramer+ population was seen in ex vivo and amplified tetramer assay.
  • Table 9 Comparison of T-cell response magnitudes calculated from ex vivo and amplified tetramer assays.
  • T-cell responses as measured by the assays described above were evaluated for all peptides contained in IMA for 28 patients at the different time points of the study.
  • a patient was scored "responsive", i.d. showed a vaccine induced immune response, if one of the blood samples taken at an after vaccination time point contained teramer-positive lymphocytes or secreted IFN- ⁇ upon stimulation with one of the peptides.
  • CDK6 PLSTIRE
  • CDK4 PSK- J3 are a distinct subset of the cyclin-dependent kinases that associate with cyclin Dl. Oncogene 9:71-79 (1994).
  • Bottaro DP Bottaro DP, Rubin JS, Faletto DL, Chan AM, Kmiecik TE, Vande Woude GF, and Aaronson SA. Identification of the hepatocyte growth factor receptor as the c-met proto-oncogene product. Science 251 :802-804 (1991).
  • MMPs matrix metalloproteinases
  • TIMPs tissue inhibitors of the matrix metalloproteinases
  • the epithelial tumor antigen MUCl is expressed in hematological malignancies and is recognized by MUCl -specific cytotoxic T-lymphocytes. Cancer Res. 61 :6846-6850 (2001).
  • Hepatocyte growth factor/scatter factor-induced activation of MEK and POK signal pathways contributes to expression of proangiogenic cytokines interleukin-8 and vascular endothelial growth factor in head and neck squamous cell carcinoma. Cancer Res. 61 :5911-5918 (2001).
  • Apolipoprotein L a new human high density lipoprotein apolipoprotein expressed by the pancreas. Identification, cloning, characterization, and plasma distribution of apolipoprotein L. J. Biol. Chem. 272:25576-25582 (1997).
  • Furge KA, Zhang YW, and Vande Woude GF Met receptor tyrosine kinase: enhanced signaling through adapter proteins. Oncogene 19:5582-5589 (2000).
  • Halaban R Melanoma cell autonomous growth: the Rb/E2F pathway. Cancer Metastasis Rev. 18:333-343 (1999).
  • Adipophilin is a specific marker of lipid accumulation in diverse cell types and diseases. Cell Tissue Res. 294:309-321 (1998).
  • Nitric oxide-mediated promotion of mammary tumour cell migration requires sequential activation of nitric oxide synthase, guanylate cyclase and mitogen-activated protein kinase. Int. J. Cancer 106:496-504 (2003).
  • HGF Hepatocyte growth factor
  • Ponzetto C Bardelli A, Maina F, Longati P, Panayotou G, Dhand R, Waterfield MD, and Comoglio PM.
  • a novel recognition motif for phosphatidylinositol 3-kinase binding mediates its association with the hepatocyte growth factor/scatter factor receptor. MoI. Cell Biol. 13:4600-4608 (1993).
  • Rae FK Rae FK
  • Stephenson SA Stephenson SA
  • Nicol DL Novel association of a diverse range of genes with renal cell carcinoma as identified by differential display.
  • HGF/SF hepatocyte growth factor/scatter factor
  • cMET HGF/SF receptor
  • Troussard X vet-Loiseau H, Macro M, Mellerin MP, Malet M, Roussel M, and Sola B. Cyclin Dl expression in patients with multiple myeloma. Hematol. J. 1 :181-185 (2000).
  • Vasef MA Brynes RK, Sturm M, Bromley C, and Robinson RA.
  • cytotoxic T-lymphocyte (CTL) response to cytomegalovirus is dominated by structural protein pp65: frequency, specificity, and T-cell receptor usage of pp65-specific CTL. J Virol. 70:7569-7579 (1996).
  • BNIP -2 induces cell elongation and membrane protrusions by interacting with Cdc42 via a unique Cdc42-binding motif within its BNIP-2 and Cdc42GAP homology domain. Exp. Cell Res. 303:263-274 (2005).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Toxicology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Genetics & Genomics (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
PCT/EP2006/008641 2005-09-05 2006-09-05 Tumor-associated peptides binding to human leukocyte antigen (hla) class i or ii molecules and related anti-cancer vaccine WO2007028573A1 (en)

Priority Applications (18)

Application Number Priority Date Filing Date Title
US12/065,725 US20090274714A1 (en) 2005-09-05 2006-09-05 Tumour-associated peptides binding to human leukocyte antigen (hla) class i or ii molecules and related anti-cancer vaccine
JP2008528437A JP5132561B2 (ja) 2005-09-05 2006-09-05 ヒト白血球抗原(hla)クラスiまたはii分子に結合する腫瘍関連ペプチドおよび関連する抗癌ワクチン
BRPI0615466-2A BRPI0615466A2 (pt) 2005-09-05 2006-09-05 peptìdios associados a tumores que se ligam a moléculas de classe i ou ii do antìgeno leucocitário humano (hla) e correspondente vacina anticáncer
CA2621414A CA2621414C (en) 2005-09-05 2006-09-05 Mmp-7 and adf derived tumour-associated peptides binding to human leukocyte antigen (hla) class i or ii molecules
NZ565956A NZ565956A (en) 2005-09-05 2006-09-05 Tumor-associated peptides binding to human leukocyte antigen (HLA) class I or II molecules and related anti-cancer vaccine
SI200630949T SI1922334T1 (sl) 2005-09-05 2006-09-05 S tumorjem povezani peptidi, ki se veĹľejo na molekule antigena humanih levkocitov (HLA), razreda I in II, ter ustrezno cepivo proti raku
KR1020087008126A KR101150663B1 (ko) 2005-09-05 2006-09-05 인간 백혈구 항원 (hla) ⅰ형 또는 ⅱ형 분자에 결합하는 종양 관련 펩티드 및 이와 관련된 항암 백신
UAA200803781A UA97095C2 (ru) 2005-09-05 2006-09-05 Противораковая вакцина, которая содержит опухолевоассоциированный пептид, который связывается с молекулами лейкоцитарного антигена человека (hla) класса i или ii, и ее применение в производстве лекарственного средства
EA200800676A EA013466B1 (ru) 2005-09-05 2006-09-05 Опухолеассоциированные пептиды, связывающиеся с молекулами человеческого лейкоцитарного антигена (hla) i или ii класса, и относящаяся к ним противораковая вакцина
AU2006289289A AU2006289289B2 (en) 2005-09-05 2006-09-05 Tumor-associated peptides binding to human leukocyte antigen (HLA) class I or II molecules and related anti-cancer vaccine
CN200680038407.7A CN101287755B (zh) 2005-09-05 2006-09-05 结合于人类白细胞抗原(hla)ⅰ类或ⅱ类分子的肿瘤相关肽及相关的抗癌疫苗
DK06777167.5T DK1922334T3 (da) 2005-09-05 2006-09-05 Tumorassocierede peptider, der binder til humant leukocyt antigen (HLA) klasse I eller II molekyler og dertil hørende anti-cancervaccine
DE602006019446T DE602006019446D1 (de) 2005-09-05 2006-09-05 An humane leukozytenantigen-(hla-)klasse-i- oder ii-moleküle bindende tumorassoziierte peptide und damit im zusammenhang stehender impfstoff gegen krebs
EP06777167A EP1922334B1 (en) 2005-09-05 2006-09-05 Tumor-associated peptides binding to human leukocyte antigen (hla) class i or ii molecules and related anti-cancer vaccine
PL06777167T PL1922334T3 (pl) 2005-09-05 2006-09-05 Peptydy towarzyszące nowotworom, wiążące się z cząsteczkami klasy I i II antygenów HLA i związana z nimi szczepionka przeciwnowotworowa
AT06777167T ATE494303T1 (de) 2005-09-05 2006-09-05 An humane leukozytenantigen-(hla-)klasse-i- oder ii-moleküle bindende tumorassoziierte peptide und damit im zusammenhang stehender impfstoff gegen krebs
NO20081690A NO20081690L (no) 2005-09-05 2008-04-04 Tumorassosierte peptider som bindes til humant leukocyttantigen (HLA) klasse I- eller II-molekyler og relatert vaksine mot kreft
HR20110240T HRP20110240T1 (en) 2005-09-05 2011-04-04 Tumor-associated peptides binding to human leukocyte antigen (hla) class i or ii molecules and related anti-cancer vaccine

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP05019255A EP1760089B1 (en) 2005-09-05 2005-09-05 Tumor-associated peptides binding to human leukocyte antigen (HLA) class I or II molecules and related anti-cancer vaccine
EP05019255.8 2005-09-05

Publications (2)

Publication Number Publication Date
WO2007028573A1 true WO2007028573A1 (en) 2007-03-15
WO2007028573A9 WO2007028573A9 (en) 2008-05-02

Family

ID=35149455

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2006/008641 WO2007028573A1 (en) 2005-09-05 2006-09-05 Tumor-associated peptides binding to human leukocyte antigen (hla) class i or ii molecules and related anti-cancer vaccine

Country Status (23)

Country Link
US (1) US20090274714A1 (ru)
EP (2) EP1760089B1 (ru)
JP (1) JP5132561B2 (ru)
KR (1) KR101150663B1 (ru)
CN (1) CN101287755B (ru)
AT (2) ATE440107T1 (ru)
AU (1) AU2006289289B2 (ru)
BR (1) BRPI0615466A2 (ru)
CA (1) CA2621414C (ru)
CY (2) CY1110536T1 (ru)
DE (2) DE602005016112D1 (ru)
DK (2) DK1760089T3 (ru)
EA (1) EA013466B1 (ru)
ES (2) ES2330013T3 (ru)
HR (1) HRP20110240T1 (ru)
NO (1) NO20081690L (ru)
NZ (1) NZ565956A (ru)
PL (2) PL1760089T3 (ru)
PT (2) PT1760089E (ru)
RS (1) RS51893B (ru)
SI (2) SI1760089T1 (ru)
UA (2) UA105210C2 (ru)
WO (1) WO2007028573A1 (ru)

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2321680A1 (es) * 2007-04-26 2009-06-09 Fundacion Para La Investigacion Biosanitaria De Andalucia Oriental - Alejandro Otero (Fibao) Restauracion de las moleculas hla de clase i mediante terapia genica empleando vectores adenovirales portando el gen de la beta 2-microglobulina.
EP2111867A1 (en) 2008-04-24 2009-10-28 Immatics Biotechnologies GmbH Novel formulations of tumour-associated peptides binding to human leukocyte antigen (HLA) class I or II molecules for vaccines
EP2119726A1 (en) 2008-05-14 2009-11-18 Immatics Biotechnologies GmbH Novel and powerful MHC-class II peptides derived from survivin and neurocan
JP2010534627A (ja) * 2007-07-27 2010-11-11 イマティクス バイオテクノロジーズ ゲーエムベーハー 腫瘍関連ペプチドおよび関連抗癌ワクチンの組成物
US20120027684A1 (en) * 2010-06-02 2012-02-02 Immatics Biotechnologies Gmbh Cancer therapy based on tumor associated antigens derived from cyclin d1
JP2012504393A (ja) * 2008-10-01 2012-02-23 イマティクス バイオテクノロジーズ ゲーエムベーハー 神経細胞性脳腫瘍など数種の腫瘍に対する新規免疫療法
JP2013028621A (ja) * 2008-04-24 2013-02-07 Immatics Biotechnologies Gmbh ワクチン用のヒト白血球抗原(hla)クラスiまたはii分子に結合する腫瘍関連ペプチドの新規組成物
JP2014138588A (ja) * 2007-07-27 2014-07-31 Immatics Biotechnologies Gmbh 免疫療法用の新規免疫原性エピトープ
US9175040B2 (en) 2007-07-27 2015-11-03 Immatics Biotechnologies Gmbh Immunotherapeutic peptides against neuronal and brain tumors and related methods and compositions
WO2017001491A2 (en) * 2015-07-01 2017-01-05 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
WO2017009400A1 (en) * 2015-07-15 2017-01-19 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against epithelial ovarian cancer and other cancers
WO2017005733A3 (en) * 2015-07-06 2017-02-16 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
EP3167898A1 (en) 2015-11-11 2017-05-17 IEO - Istituto Europeo di Oncologia Srl Method for obtaining tumor peptides and uses thereof
CN107912043A (zh) * 2015-07-06 2018-04-13 伊玛提克斯生物技术有限公司 用于食管癌和其他癌症免疫治疗的新型肽和肽组合物
WO2018104407A1 (en) 2016-12-08 2018-06-14 Immatics Biotechnologies Gmbh T cell receptors with improved pairing
US10900072B2 (en) 2012-06-27 2021-01-26 Rutgers, The State University Of New Jersey Rapid assays for T-cell activation by RNA measurements using flow cytometry
AU2017315139B2 (en) * 2016-08-26 2021-11-11 Immatics Biotechnologies Gmbh Novel peptides and scaffolds for use in immunotherapy against head and neck squamous cell carcinoma and other cancers
EP3919507A3 (en) * 2015-07-01 2022-01-12 Immatics Biotechnologies GmbH Novel peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
WO2022226535A1 (en) * 2021-04-23 2022-10-27 Flow Pharma Inc. Vaccine for sars-cov-2
US11679147B2 (en) 2014-12-23 2023-06-20 Immatics Biotechnologies Gmbh Method of eliciting a CD8+ cytotoxic response in hepatocellular carcinoma patients with a population of activated T cells
US11786583B2 (en) 2014-12-23 2023-10-17 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against Hepatocellular carcinoma (HCC) and other cancers
EP4317432A2 (en) 2016-12-08 2024-02-07 Immatics Biotechnologies GmbH T cell receptors with improved pairing
US11999774B2 (en) 2015-07-06 2024-06-04 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers

Families Citing this family (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE461214T1 (de) * 2005-09-05 2010-04-15 Immatics Biotechnologies Gmbh Tumor-assoziierte peptide, welche an unterschiedliche menschliche leukozytenantigene der klasse ii binden
US20090004213A1 (en) 2007-03-26 2009-01-01 Immatics Biotechnologies Gmbh Combination therapy using active immunotherapy
AU2016204711B2 (en) * 2008-10-01 2018-03-01 Immatics Biotechnologies Gmbh Novel immunotherapy against several tumors including neuronal and brain tumors
GB201004575D0 (en) * 2010-03-19 2010-05-05 Immatics Biotechnologies Gmbh Composition of tumor associated peptides and related anti cancer vaccine for the treatment of gastric cancer and other cancers
GB201004551D0 (en) * 2010-03-19 2010-05-05 Immatics Biotechnologies Gmbh NOvel immunotherapy against several tumors including gastrointestinal and gastric cancer
GB201006360D0 (en) * 2010-04-16 2010-06-02 Immatics Biotechnologies Gmbh Method for differentially quantifying naturally processed HLA-restricted peptides for cancer, autoimmune and infectious diseases immunotherapy development
JP6064186B2 (ja) * 2010-12-02 2017-01-25 オンコセラピー・サイエンス株式会社 Tomm34ペプチドおよびそれを含むワクチン
WO2013005164A2 (en) * 2011-07-05 2013-01-10 Cadila Pharmaceuticals Limited Cancer antigen
MY170949A (en) * 2011-07-11 2019-09-20 Cancer Res Initiatives Foundation A peptide capable of binding with a human leukocyte antigen (hla) molecule, a cancer vaccine derived from said peptide and use of said cancer vaccine
US20140017267A1 (en) * 2012-07-13 2014-01-16 Matthias W Rath Metalloproteinase oligopeptides and their therapeutic use
TWI777197B (zh) * 2013-08-05 2022-09-11 德商伊瑪提克斯生物科技有限公司 新穎肽類,細胞及其用於治療多種腫瘤的用途,其製造方法及包含其等之醫藥組成物(六)
EP3369432A1 (en) 2013-12-09 2018-09-05 Targovax Asa A peptide mixture
CR20210232A (es) 2015-03-17 2022-06-24 Immatics Biotechnologies Gmbh NUEVOS PÉPTIDOS Y NUEVA COMBINACIÓN DE PÉPTIDOS PARA EL USO EN LA INMUNOTERAPIA CONTRA EL CÁNCER DE PÁNCREAS Y OTROS TIPOS DE CÁNCER (Divisional 2017-0419)
GB201504502D0 (en) * 2015-03-17 2015-04-29 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against pancreatic cancer and other cancers
GB201505305D0 (en) 2015-03-27 2015-05-13 Immatics Biotechnologies Gmbh Novel Peptides and combination of peptides for use in immunotherapy against various tumors
DK3273986T3 (da) 2015-03-27 2024-02-12 Immatics Biotechnologies Gmbh Hidtil ukendte peptider og kombination af peptider til anvendelse ved immunterapi mod forskellige tumorer
GB201505585D0 (en) * 2015-03-31 2015-05-13 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides and scaffolds for use in immunotherapy against renal cell carinoma (RCC) and other cancers
GB201507030D0 (en) * 2015-04-24 2015-06-10 Immatics Biotechnologies Gmbh Immunotherapy against lung cancers, in particular NSCLC
GB201510771D0 (en) 2015-06-19 2015-08-05 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy and methods for generating scaffolds for the use against pancreatic cancer
TWI751557B (zh) 2015-06-19 2022-01-01 德商英麥提克生物技術股份有限公司 用於免疫治療的新穎胜肽及胜肽組合物與產生用於抗胰臟癌及其他癌症的支架的方法
ZA201707555B (en) 2015-06-25 2018-11-28 Immatics Biotechnologies Gmbh Novel cell epitopes and combination of cell epitopes for use in the immuno-therapy of myeloma and other cancers
GB201511191D0 (en) 2015-06-25 2015-08-12 Immatics Biotechnologies Gmbh T-cell epitopes for the immunotherapy of myeloma
GB201517538D0 (en) 2015-10-05 2015-11-18 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against small cell lung cancer and other cancers
MY198087A (en) 2015-10-05 2023-07-31 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against small cell lung cancer and other cancers
US20190070275A1 (en) * 2016-02-22 2019-03-07 Oceanside Biotechnology Neoantigen compositions and methods of using the same in immunooncotherapy
MA44524A (fr) * 2016-03-31 2019-02-06 Neon Therapeutics Inc Néoantigènes et leurs procédés d'utilisation
CN106986932A (zh) * 2017-04-06 2017-07-28 海口市人民医院 一种c‑Met表位肽及其应用
WO2019160099A1 (ja) * 2018-02-15 2019-08-22 大塚製薬株式会社 がん抗原ペプチド
CN110804642B (zh) * 2019-09-10 2022-06-21 清华大学 Gpr174免疫学功能及其用途
EP4061424A4 (en) * 2019-11-14 2023-01-25 Bogazici Universitesi ASC STAINS USED IN CANCER IMMUNOTHERAPY
CN114524870A (zh) * 2020-11-23 2022-05-24 香雪生命科学技术(广东)有限公司 源自于ssx2抗原的短肽
CN114478711A (zh) * 2022-01-05 2022-05-13 成都朗谷生物科技股份有限公司 一种针对乙型肝炎病毒的抗原肽及其应用

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000006723A1 (en) * 1998-07-30 2000-02-10 Yeda Research And Development Company Ltd At The Weizmann Institute Of Science Tumor associated antigen peptides and use of same as anti-tumor vaccines
US6096313A (en) * 1996-02-09 2000-08-01 Ludwig Institute For Cancer Research Compositions containing immunogenic molecules and granulocyte-macrophage colony stimulating factor, as an adjuvant
US20040171543A1 (en) * 2002-08-13 2004-09-02 Anne-Claude Dublanchet Matrix metalloproteinase inhibitors and methods for identification of lead compounds

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4440859A (en) 1977-05-27 1984-04-03 The Regents Of The University Of California Method for producing recombinant bacterial plasmids containing the coding sequences of higher organisms
US4704362A (en) 1977-11-08 1987-11-03 Genentech, Inc. Recombinant cloning vehicle microbial polypeptide expression
YU44186B (en) 1978-12-22 1990-04-30 Biogen Nv Process for obtaining recombinant dnk molecules
US4530901A (en) 1980-01-08 1985-07-23 Biogen N.V. Recombinant DNA molecules and their use in producing human interferon-like polypeptides
US4678751A (en) 1981-09-25 1987-07-07 Genentech, Inc. Hybrid human leukocyte interferons
US4766075A (en) 1982-07-14 1988-08-23 Genentech, Inc. Human tissue plasminogen activator
US4582800A (en) 1982-07-12 1986-04-15 Hoffmann-La Roche Inc. Novel vectors and method for controlling interferon expression
US4757006A (en) 1983-10-28 1988-07-12 Genetics Institute, Inc. Human factor VIII:C gene and recombinant methods for production
US4677063A (en) 1985-05-02 1987-06-30 Cetus Corporation Human tumor necrosis factor
US4810648A (en) 1986-01-08 1989-03-07 Rhone Poulenc Agrochimie Haloarylnitrile degrading gene, its use, and cells containing the gene
EP0671926B1 (en) * 1992-08-11 2002-11-13 President And Fellows Of Harvard College Immunomodulatory peptides
AUPM322393A0 (en) 1993-12-24 1994-01-27 Austin Research Institute, The Mucin carbohydrate compounds and their use in immunotherapy
NZ290089A (en) 1994-07-27 1999-05-28 Queensland Inst Med Res Recombinant polyepitope cytotoxic t lymphocyte (ctl) vaccines
ATE244300T1 (de) 1996-01-17 2003-07-15 Imp College Innovations Ltd Immunotherapie mit verwendung von zytotoxischen t lymphozyten (ctl)
US6316213B1 (en) * 1997-03-19 2001-11-13 The Board Of Trustees Of The University Of Arkansas Methods for the early diagnosis of ovarian, breast and lung cancer
DE19917195B4 (de) 1999-04-16 2006-09-28 Immatics Biotechnologies Gmbh Peptid zur Auslösung einer Immunreaktion gegen Tumorzellen, diese enthaltende pharmzeutische Zusammensetzungen, dessen Verwendungen, dafür codierende Nukleinsäure und diese Nukleinsäure enthaltender Expressionsvektor
KR100358068B1 (ko) * 1999-12-28 2002-10-25 주식회사 하이닉스반도체 플래쉬 메모리 소자의 제조방법
US20060045881A1 (en) * 2004-08-26 2006-03-02 Board Of Regents, The University Of Texas System Anti-cancer vaccines

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6096313A (en) * 1996-02-09 2000-08-01 Ludwig Institute For Cancer Research Compositions containing immunogenic molecules and granulocyte-macrophage colony stimulating factor, as an adjuvant
WO2000006723A1 (en) * 1998-07-30 2000-02-10 Yeda Research And Development Company Ltd At The Weizmann Institute Of Science Tumor associated antigen peptides and use of same as anti-tumor vaccines
US20040171543A1 (en) * 2002-08-13 2004-09-02 Anne-Claude Dublanchet Matrix metalloproteinase inhibitors and methods for identification of lead compounds

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
BARNETT J ET AL.: "Production, purification and characterization of human matrilysin (PUMP) from recombinant chinese hamster ovary cells", PROTEIN EXPRESSION AND PURIFICATION, vol. 5, 1994, pages 27 - 36, XP002352893 *
BROSSART P ET AL: "Identification of HLA-A2-restricted T-cell epitopes derived from the MUC1 tumor antigen for broadly applicable vaccine therapies", BLOOD, W.B.SAUNDERS COMPANY, ORLANDO, FL, US, vol. 93, no. 12, 15 June 1999 (1999-06-15), pages 4309 - 4317, XP002147432, ISSN: 0006-4971 *
BROSSART P ET AL: "Virus-mediated delivery of antigenic peptides into dendritic cells as a means to induce CTL", JOURNAL OF IMMUNOLOGY, THE WILLIAMS AND WILKINS CO. BALTIMORE, US, vol. 7, no. 158, 1 April 1997 (1997-04-01), pages 3270 - 3276, XP002075947, ISSN: 0022-1767 *
HEID H W ET AL: "Adipocyte differentiation-related protein is secreted into milk as a constituent of milk lipid globule membrane", BIOCHEMICAL JOURNAL, PORTLAND PRESS, LONDON, GB, vol. 320, no. PART 3, 15 December 1996 (1996-12-15), pages 1025 - 1030, XP002060680, ISSN: 0264-6021 *
SCHMIDT SM ET AL: "Induction of adipophilin-specific cytotoxic T lymphocytes using a novel HLA-A2 binding peptide that mediates tumor cell lysis", CANCER RESEARCH, vol. 64, 2004, pages 1164 - 1170, XP002352895 *
SUMI TOSHIYUKI ET AL: "Expression of matrix metalloproteinases 7 and 2 in human renal cell carcinoma", ONCOLOGY REPORTS, NATIONAL HELLENIC RESEARCH FOUNDATION, ATHENS, GR, vol. 10, no. 3, May 2003 (2003-05-01), pages 567 - 570, XP002352032, ISSN: 1021-335X *
WEINSCHENK T ET AL: "Integrated functional genomics approach for the design of patient-individual antitumor vaccines", CANCER RESEARCH, AMERICAN ASSOCIATION FOR CANCER RESEARCH, BALTIMORE, MD, US, vol. 62, no. 20, 15 October 2002 (2002-10-15), pages 5818 - 5827, XP002266492, ISSN: 0008-5472 *
WELCH AR ET AL.: "Purification of human matrilysin produced in Escherichia coli and characterization using a new optimized fluorogenic peptide substrate", ARCHIVES OF BIOCHEMISTRY AND BIOPHYSICS, vol. 324, no. 1, 1995, pages 59 - 64, XP002352894 *

Cited By (106)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2321680A1 (es) * 2007-04-26 2009-06-09 Fundacion Para La Investigacion Biosanitaria De Andalucia Oriental - Alejandro Otero (Fibao) Restauracion de las moleculas hla de clase i mediante terapia genica empleando vectores adenovirales portando el gen de la beta 2-microglobulina.
US9175040B2 (en) 2007-07-27 2015-11-03 Immatics Biotechnologies Gmbh Immunotherapeutic peptides against neuronal and brain tumors and related methods and compositions
US10420800B2 (en) 2007-07-27 2019-09-24 Immatics Biotechnologies Gmbh Immunotherapy against neuronal and brain tumors
US11986497B2 (en) 2007-07-27 2024-05-21 Immatics Biotechnologies Gmbh Immunotherapy against neuronal and brain tumors
US11648276B2 (en) 2007-07-27 2023-05-16 Immatics Biotechnologies Gmbh Immunotherapy against neuronal and brain tumors
JP2010534627A (ja) * 2007-07-27 2010-11-11 イマティクス バイオテクノロジーズ ゲーエムベーハー 腫瘍関連ペプチドおよび関連抗癌ワクチンの組成物
US10709735B2 (en) 2007-07-27 2020-07-14 Immatics Biotechnologies Gmbh Immunotherapy against neuronal and brain tumors
US10434120B2 (en) 2007-07-27 2019-10-08 Immatics Biotechnologies Gmbh Immunotherapy against neuronal and brain tumors
JP2014138588A (ja) * 2007-07-27 2014-07-31 Immatics Biotechnologies Gmbh 免疫療法用の新規免疫原性エピトープ
JP2013028621A (ja) * 2008-04-24 2013-02-07 Immatics Biotechnologies Gmbh ワクチン用のヒト白血球抗原(hla)クラスiまたはii分子に結合する腫瘍関連ペプチドの新規組成物
CN105749285A (zh) * 2008-04-24 2016-07-13 伊玛提克斯生物技术有限公司 结合到人类白细胞抗原(hla)ⅰ类或ⅱ类分子上用作疫苗的肿瘤相关肽新制剂
EP2111867A1 (en) 2008-04-24 2009-10-28 Immatics Biotechnologies GmbH Novel formulations of tumour-associated peptides binding to human leukocyte antigen (HLA) class I or II molecules for vaccines
US9289478B2 (en) 2008-04-24 2016-03-22 Immatics Biotechnologies Gmbh Formulations of tumour-associated peptides binding to human leukocyte antigen (HLA) class I or class II molecules for vaccine
JP2009292810A (ja) * 2008-04-24 2009-12-17 Immatics Biotechnologies Gmbh ワクチン用のヒト白血球抗原(hla)クラスiまたはii分子に結合する腫瘍関連ペプチドの新規組成物
EA017492B1 (ru) * 2008-04-24 2012-12-28 Имматикс Байотекнолоджиз Гмбх Новые композиции опухолеассоциированных пептидов, связывающихся с молекулами человеческого лейкоцитарного антигена (hla) i или ii класса, для вакцин
US9283267B2 (en) 2008-04-24 2016-03-15 Immatics Biotechnologies Gmbh Formulations of tumour-associated peptides binding to human leukocyte antigen (HLA) class I or class II molecules for vaccines
EP2113253A1 (en) 2008-04-30 2009-11-04 Immatics Biotechnologies GmbH Novel formulations of tumour-associated peptides binding to human leukocyte antigen (HLA) class I or II molecules for vaccines
JP2011520436A (ja) * 2008-05-14 2011-07-21 イマティクス バイオテクノロジーズ ゲーエムベーハー サービビン由来の新規で強力なmhcクラスiiペプチド
EP2899206A1 (en) 2008-05-14 2015-07-29 immatics biotechnologies GmbH Novel and powerful MHC-class II peptides derived from survivin
EP2119726A1 (en) 2008-05-14 2009-11-18 Immatics Biotechnologies GmbH Novel and powerful MHC-class II peptides derived from survivin and neurocan
JP2012504393A (ja) * 2008-10-01 2012-02-23 イマティクス バイオテクノロジーズ ゲーエムベーハー 神経細胞性脳腫瘍など数種の腫瘍に対する新規免疫療法
JP2017029135A (ja) * 2008-10-01 2017-02-09 イマティクス バイオテクノロジーズ ゲーエムベーハー 神経細胞性脳腫瘍など数種の腫瘍に対する新規免疫療法
JP2017000148A (ja) * 2008-10-01 2017-01-05 イマティクス バイオテクノロジーズ ゲーエムベーハー 神経細胞性脳腫瘍など数種の腫瘍に対する新規免疫療法
JP2014239681A (ja) * 2008-10-01 2014-12-25 イマティクス バイオテクノロジーズ ゲーエムベーハー 神経細胞性脳腫瘍など数種の腫瘍に対する新規免疫療法
JP2017018101A (ja) * 2008-10-01 2017-01-26 イマティクス バイオテクノロジーズ ゲーエムベーハー 神経細胞性脳腫瘍など数種の腫瘍に対する新規免疫療法
JP2017018102A (ja) * 2008-10-01 2017-01-26 イマティクス バイオテクノロジーズ ゲーエムベーハー 神経細胞性脳腫瘍など数種の腫瘍に対する新規免疫療法
JP2017023136A (ja) * 2008-10-01 2017-02-02 イマティクス バイオテクノロジーズ ゲーエムベーハー 神経細胞性脳腫瘍など数種の腫瘍に対する新規免疫療法
JP2016145210A (ja) * 2008-10-01 2016-08-12 イマティクス バイオテクノロジーズ ゲーエムベーハー 神経細胞性脳腫瘍など数種の腫瘍に対する新規免疫療法
JP2016047825A (ja) * 2008-10-01 2016-04-07 イマティクス バイオテクノロジーズ ゲーエムベーハー 神経細胞性脳腫瘍など数種の腫瘍に対する新規免疫療法
US9056069B2 (en) * 2010-06-02 2015-06-16 Immatics Biotechnologies Gmbh Cancer therapy based on tumor associated antigens derived from cyclin D1
US20120027684A1 (en) * 2010-06-02 2012-02-02 Immatics Biotechnologies Gmbh Cancer therapy based on tumor associated antigens derived from cyclin d1
US11767551B2 (en) 2012-06-27 2023-09-26 Rutgers, The State University Of New Jersey Rapid assays for T-cell activation by RNA measurements using flow cytometry
US10900072B2 (en) 2012-06-27 2021-01-26 Rutgers, The State University Of New Jersey Rapid assays for T-cell activation by RNA measurements using flow cytometry
US11679147B2 (en) 2014-12-23 2023-06-20 Immatics Biotechnologies Gmbh Method of eliciting a CD8+ cytotoxic response in hepatocellular carcinoma patients with a population of activated T cells
US11786583B2 (en) 2014-12-23 2023-10-17 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against Hepatocellular carcinoma (HCC) and other cancers
US10472401B2 (en) 2015-07-01 2019-11-12 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
US10738094B2 (en) 2015-07-01 2020-08-11 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
WO2017001491A2 (en) * 2015-07-01 2017-01-05 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
US11912748B2 (en) 2015-07-01 2024-02-27 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
US11912749B2 (en) 2015-07-01 2024-02-27 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
WO2017001491A3 (en) * 2015-07-01 2017-03-30 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
US10227388B2 (en) 2015-07-01 2019-03-12 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against non-small cell lung cancer and other cancers
US10239925B2 (en) 2015-07-01 2019-03-26 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
US11485765B2 (en) 2015-07-01 2022-11-01 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
US10253076B2 (en) 2015-07-01 2019-04-09 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
US11401310B2 (en) 2015-07-01 2022-08-02 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
US10280205B2 (en) 2015-07-01 2019-05-07 Immatics Biotechnologies Gmbh Method for treating cancer with an activated T lymphocyte that selectively recognizes a cancer cell consisting of specific peptide
US11384129B2 (en) 2015-07-01 2022-07-12 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
US11384128B2 (en) 2015-07-01 2022-07-12 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
US11384127B2 (en) 2015-07-01 2022-07-12 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
US11230581B2 (en) 2015-07-01 2022-01-25 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
EP3919507A3 (en) * 2015-07-01 2022-01-12 Immatics Biotechnologies GmbH Novel peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
US10464978B2 (en) 2015-07-01 2019-11-05 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
US10934333B2 (en) 2015-07-01 2021-03-02 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
US10174089B2 (en) 2015-07-01 2019-01-08 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
US10494413B2 (en) 2015-07-01 2019-12-03 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
US10533041B2 (en) 2015-07-01 2020-01-14 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
EP3712165A3 (en) * 2015-07-01 2020-12-02 immatics biotechnologies GmbH Novel peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
US10487132B2 (en) 2015-07-06 2019-11-26 Inmatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
WO2017005733A3 (en) * 2015-07-06 2017-02-16 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
US11999774B2 (en) 2015-07-06 2024-06-04 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
US10703795B2 (en) 2015-07-06 2020-07-07 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
CN107912043A (zh) * 2015-07-06 2018-04-13 伊玛提克斯生物技术有限公司 用于食管癌和其他癌症免疫治疗的新型肽和肽组合物
US10189886B2 (en) 2015-07-06 2019-01-29 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
US10626162B2 (en) 2015-07-06 2020-04-21 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
US11939366B2 (en) 2015-07-06 2024-03-26 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
US10829537B2 (en) 2015-07-06 2020-11-10 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
EP3736283A1 (en) * 2015-07-06 2020-11-11 immatics biotechnologies GmbH Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
US11932678B2 (en) 2015-07-06 2024-03-19 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
US10202437B2 (en) 2015-07-06 2019-02-12 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
US10214577B2 (en) 2015-07-06 2019-02-26 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
US11359003B2 (en) 2015-07-06 2022-06-14 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
US10221229B2 (en) 2015-07-06 2019-03-05 Inmatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
AU2016289754B2 (en) * 2015-07-06 2021-02-18 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
US11608370B2 (en) 2015-07-06 2023-03-21 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
US10239930B2 (en) 2015-07-06 2019-03-26 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
US11401319B2 (en) 2015-07-06 2022-08-02 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
US10273282B2 (en) 2015-07-06 2019-04-30 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
US10294288B2 (en) 2015-07-06 2019-05-21 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
US10364282B2 (en) 2015-07-06 2019-07-30 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
CN107912043B (zh) * 2015-07-06 2022-02-18 伊玛提克斯生物技术有限公司 用于食管癌和其他癌症免疫治疗的新型肽和肽组合物
US10167331B1 (en) 2015-07-06 2019-01-01 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
TWI765857B (zh) * 2015-07-06 2022-06-01 德商英麥提克生物技術股份有限公司 用於對抗食道癌及其他癌症的新穎胜肽及胜肽的組合物
US11278571B2 (en) 2015-07-15 2022-03-22 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against epithelial ovarian cancer and other cancers
US10568909B2 (en) 2015-07-15 2020-02-25 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against epithelial ovarian cancer and other cancers
US11246889B2 (en) 2015-07-15 2022-02-15 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against epithelial ovarian cancer and other cancers
US10639332B2 (en) 2015-07-15 2020-05-05 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against epithelial ovarian cancer and other cancers
US10463696B2 (en) 2015-07-15 2019-11-05 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against epithelial ovarian cancer and other cancers
US10869897B2 (en) 2015-07-15 2020-12-22 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against epithelial ovarian cancer and other cancers
US10722538B2 (en) 2015-07-15 2020-07-28 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against epithelial ovarian cancer and other cancers
US11071756B2 (en) 2015-07-15 2021-07-27 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against epithelial ovarian cancer and other cancers
EP3705491A1 (en) * 2015-07-15 2020-09-09 Immatics Biotechnologies GmbH Novel peptides and combination of peptides for use in immunotherapy against epithelial ovarian cancer and other cancers
US10881690B2 (en) 2015-07-15 2021-01-05 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against epithelial ovarian cancer and other cancers
IL256697A (en) * 2015-07-15 2018-03-29 Immatics Biotechnologies Gmbh New peptides and a combination of peptides for use in immunotherapy against ovarian cancer and other cancers
US10888587B2 (en) 2015-07-15 2021-01-12 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against epithelial ovarian cancer and other cancers
WO2017009400A1 (en) * 2015-07-15 2017-01-19 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against epithelial ovarian cancer and other cancers
US11806366B2 (en) 2015-07-15 2023-11-07 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against epithelial ovarian cancer and other cancers
US11819518B2 (en) 2015-07-15 2023-11-21 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against epithelial ovarian cancer and other cancers
AU2022200745B2 (en) * 2015-07-15 2024-01-18 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against epithelial ovarian cancer and other cancers
US10639331B2 (en) 2015-07-15 2020-05-05 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against epithelial ovarian cancer and other cancers
EP3167898A1 (en) 2015-11-11 2017-05-17 IEO - Istituto Europeo di Oncologia Srl Method for obtaining tumor peptides and uses thereof
AU2017315139B2 (en) * 2016-08-26 2021-11-11 Immatics Biotechnologies Gmbh Novel peptides and scaffolds for use in immunotherapy against head and neck squamous cell carcinoma and other cancers
EP4317432A2 (en) 2016-12-08 2024-02-07 Immatics Biotechnologies GmbH T cell receptors with improved pairing
WO2018104407A1 (en) 2016-12-08 2018-06-14 Immatics Biotechnologies Gmbh T cell receptors with improved pairing
DE102016123893A1 (de) 2016-12-08 2018-06-14 Immatics Biotechnologies Gmbh T-Zellrezeptoren mit verbesserter Bindung
WO2022226535A1 (en) * 2021-04-23 2022-10-27 Flow Pharma Inc. Vaccine for sars-cov-2

Also Published As

Publication number Publication date
EP1922334B1 (en) 2011-01-05
PL1760089T3 (pl) 2010-03-31
NO20081690L (no) 2008-04-04
EA200800676A1 (ru) 2008-08-29
WO2007028573A9 (en) 2008-05-02
JP2009506762A (ja) 2009-02-19
PT1922334E (pt) 2011-04-01
EP1922334A1 (en) 2008-05-21
ES2358802T3 (es) 2011-05-13
CA2621414C (en) 2016-10-04
UA97095C2 (ru) 2012-01-10
DK1760089T3 (da) 2009-11-16
UA105210C2 (ru) 2014-04-25
SI1922334T1 (sl) 2011-04-29
NZ565956A (en) 2011-06-30
DK1922334T3 (da) 2011-04-18
AU2006289289B2 (en) 2012-04-19
PT1760089E (pt) 2009-10-19
AU2006289289A1 (en) 2007-03-15
PL1922334T3 (pl) 2011-06-30
ATE494303T1 (de) 2011-01-15
RS51893B (en) 2012-02-29
KR20080052647A (ko) 2008-06-11
BRPI0615466A2 (pt) 2011-05-17
EP1760089B1 (en) 2009-08-19
CY1113062T1 (el) 2016-04-13
DE602005016112D1 (de) 2009-10-01
ATE440107T1 (de) 2009-09-15
CY1110536T1 (el) 2015-04-29
KR101150663B1 (ko) 2012-05-25
JP5132561B2 (ja) 2013-01-30
EP1760089A1 (en) 2007-03-07
CA2621414A1 (en) 2007-03-15
ES2330013T3 (es) 2009-12-03
EA013466B1 (ru) 2010-04-30
CN101287755A (zh) 2008-10-15
CN101287755B (zh) 2014-12-31
DE602006019446D1 (de) 2011-02-17
US20090274714A1 (en) 2009-11-05
HRP20110240T1 (en) 2011-05-31
SI1760089T1 (sl) 2009-12-31

Similar Documents

Publication Publication Date Title
EP1922334B1 (en) Tumor-associated peptides binding to human leukocyte antigen (hla) class i or ii molecules and related anti-cancer vaccine
US10618945B2 (en) Tumor-associated peptides binding promiscuously to human leukocyte antigen (HLA) class II molecules
US9056069B2 (en) Cancer therapy based on tumor associated antigens derived from cyclin D1
WO2006037421A2 (en) Immunogenic t-helper epitopes from human tumour antigens and immunotherapeutic methods using said epitopes
WO2006114307A1 (en) T-cell epitopes from the oncofetal antigen-immature laminin receptor protein and medical uses thereof
JP5736333B2 (ja) 無差別的にヒト白血球抗体(hla)クラスii分子に結合する腫瘍関連ペプチド
JP5736334B2 (ja) 無差別的にヒト白血球抗体(hla)クラスii分子に結合する腫瘍関連ペプチド

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200680038407.7

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2006289289

Country of ref document: AU

Ref document number: 565956

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2006777167

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2008528437

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2621414

Country of ref document: CA

Ref document number: MX/a/2008/003157

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2006289289

Country of ref document: AU

Date of ref document: 20060905

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2006289289

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 200800676

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 1640/CHENP/2008

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 1020087008126

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2006777167

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 12065725

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: P-2011/0146

Country of ref document: RS

ENP Entry into the national phase

Ref document number: PI0615466

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20080305